You are on page 1of 72

Antithrombotic Therapy in Neonates and

Children : Antithrombotic Therapy and


Prevention of Thrombosis, 9th ed: American
College of Chest Physicians Evidence-Based
Clinical Practice Guidelines
Paul Monagle, Anthony K. C. Chan, Neil A. Goldenberg, Rebecca N.
Ichord, Janna M. Journeycake, Ulrike Nowak-Gttl and Sara K. Vesely
Chest 2012;141;e737S-e801S
DOI 10.1378/chest.11-2308
The online version of this article, along with updated information and
services can be found online on the World Wide Web at:
http://chestjournal.chestpubs.org/content/141/2_suppl/e737S.full.html
Supplemental material related to this article is available at:
http://chestjournal.chestpubs.org/content/suppl/2012/02/03/141.2_suppl.
e737S.DC1.html

Chest is the official journal of the American College of Chest


Physicians. It has been published monthly since 1935.
Copyright2012by the American College of Chest Physicians, 3300
Dundee Road, Northbrook, IL 60062. All rights reserved. No part of
this article or PDF may be reproduced or distributed without the prior
written permission of the copyright holder.
(http://chestjournal.chestpubs.org/site/misc/reprints.xhtml)
ISSN:0012-3692

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

CHEST

Supplement

ANTITHROMBOTIC THERAPY AND PREVENTION OF THROMBOSIS, 9TH ED: ACCP GUIDELINES

Antithrombotic Therapy in Neonates


and Children
Antithrombotic Therapy and Prevention of Thrombosis,
9th ed: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines
Paul Monagle, MBBS, MD, FCCP; Anthony K. C. Chan, MBBS;
Neil A. Goldenberg, MD, PhD; Rebecca N. Ichord, MD;
Janna M. Journeycake, MD, MSCS; Ulrike Nowak-Gttl, MD; and Sara K. Vesely, PhD

Background: Neonates and children differ from adults in physiology, pharmacologic responses to
drugs, epidemiology, and long-term consequences of thrombosis. This guideline addresses optimal strategies for the management of thrombosis in neonates and children.
Methods: The methods of this guideline follow those described in the Methodology for the Development of Antithrombotic Therapy and Prevention of Thrombosis Guidelines: Antithrombotic
Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians EvidenceBased Clinical Practice Guidelines.
Results: We suggest that where possible, pediatric hematologists with experience in thromboembolism manage pediatric patients with thromboembolism (Grade 2C). When this is not possible,
we suggest a combination of a neonatologist/pediatrician and adult hematologist supported by
consultation with an experienced pediatric hematologist (Grade 2C). We suggest that therapeutic
unfractionated heparin in children is titrated to achieve a target anti-Xa range of 0.35 to 0.7 units/mL
or an activated partial thromboplastin time range that correlates to this anti-Xa range or to a
protamine titration range of 0.2 to 0.4 units/mL (Grade 2C). For neonates and children receiving
either daily or bid therapeutic low-molecular-weight heparin, we suggest that the drug be monitored to a target range of 0.5 to 1.0 units/mL in a sample taken 4 to 6 h after subcutaneous injection or, alternatively, 0.5 to 0.8 units/mL in a sample taken 2 to 6 h after subcutaneous injection
(Grade 2C).
Conclusions: The evidence supporting most recommendations for antithrombotic therapy in
neonates and children remains weak. Studies addressing appropriate drug target ranges and
monitoring requirements are urgently required in addition to site- and clinical situation-specific
thrombosis management strategies.
CHEST 2012; 141(2)(Suppl):e737Se801S
Abbreviations: ACCP 5 American College of Chest Physicians; AIS 5 arterial ischemic stroke; ALL 5 acute lymphoblastic
leukemia; APLA 5 antiphospholipid antibody; aPTT 5 activated partial thromboplastin time; ASA 5 acetylsalicyclic
acid; BCPS 5 bilateral cavopulmonary shunt; CC 5 cardiac catheterization; CSVT 5 cerebral sinovenous thrombosis;
CVAD 5 central venous access device; FFP 5 fresh frozen plasma; HIT 5 heparin-induced thrombocytopenia;
ICH 5 intracerebral hemorrhage; INR 5 international normalized ratio; IVC 5 inferior vena cava; IVH 5 intraventricular
hemorrhage; LMWH 5 low-molecular-weight heparin; MBTS 5 modified Blalock-Taussig shunt; NEC 5 necrotizing
enterocolitis; PE 5 pulmonary embolism; PFA 5 platelet function analyzer; PFO 5 patent foramen ovale; PICU 5 pediatric
ICU; PTS 5 postthrombotic syndrome; RCT 5 randomized control trial; RR 5 risk ratio; rUK 5 recombinant urokinase; RVT 5 renal vein thrombosis; TCD 5 transcranial Doppler; TE 5 thromboembolism; TIA 5 transient ischemic
attack; tPA 5 tissue plasminogen activator; TPN 5 total parenteral nutrition; UAC 5 umbilical arterial catheter;
UFH 5 unfractionated heparin; UVC 5 umbilical venous catheter; VAD5 ventricular assist device; VKA 5 vitamin
K antagonist

www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e737S

Summary of Recommendations
Note on Shaded Text: Throughout this guideline,
shading is used within the summary of recommendations sections to indicate recommendations that are
newly added or have been changed since the publication of Antithrombotic and Thrombolytic Therapy:
American College of Chest Physicians EvidenceBased Clinical Practice Guidelines (8th Edition). Recommendations that remain unchanged are not shaded.
1.0. We suggest that where possible, pediatric
hematologists with experience in thromboembolism (TE) manage pediatric patients with TE
(Grade 2C). When this is not possible, we suggest
a combination of a neonatologist/pediatrician
and adult hematologist supported by consultation with an experienced pediatric hematologist
(Grade 2C).
1.1. We suggest that therapeutic unfractionated
heparin (UFH) in children is titrated to achieve
a target range of anti-Xa activity of 0.35 to
0.7 units/mL or an activated partial thromboplastin time (aPTT) range that correlates to this
anti-Xa range or to a protamine titration range
Revision accepted August 31, 2011.
Affiliations: From the Haematology Department (Dr Monagle),
The Royal Childrens Hospital, Department of Paediatrics, The
University of Melbourne, Murdoch Childrens Research Institute,
Melbourne, VIC, Australia; Department of Pediatrics (Dr Chan),
McMaster University, Hamilton, ON, Canada; Department of
Pediatrics (Dr Goldenberg), Section of Hematology/Oncology/
Bone Marrow Transplantation and Mountain States Regional
Hemophilia and Thrombosis Center, University of Colorado,
Aurora, CO; Department of Neurology (Dr Ichord), Childrens
Hospital of Philadelphia, Philadelphia, PA; Department of
Pediatrics (Dr Journeycake), University of Texas Southwestern
Medical Center at Dallas, Dallas, TX; Thrombosis and Hemostasis
Unit (Dr Nowak-Gttl), Institute of Clinical Chemistry, University Hospital Kiel, Kiel, Germany; and Department of Biostatistics
and Epidemiology (Dr Vesely), University of Oklahoma Health
Sciences Center, Oklahoma City, OK.
Funding/Support: The Antithrombotic Therapy and Prevention
of Thrombosis, 9th ed: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines received support from
the National Heart, Lung, and Blood Institute [R13 HL104758]
and Bayer Schering Pharma AG. Support in the form of educational grants was also provided by Bristol-Myers Squibb; Pfizer,
Inc; Canyon Pharmaceuticals; and sanofi-aventis US.
Disclaimer: American College of Chest Physician guidelines are
intended for general information only, are not medical advice, and
do not replace professional medical care and physician advice,
which always should be sought for any medical condition. The
complete disclaimer for this guideline can be accessed at http://
chestjournal.chestpubs.org/content/141/2_suppl/1S.
Correspondence to: Sara K. Vesely, PhD, Department of Biostatistics and Epidemiology, The University of Oklahoma Health
Sciences Center, 801 NE 13th St, CHB 309, Oklahoma City,
OK 73104; e-mail: sara-vesely@ouhsc.edu
2012 American College of Chest Physicians. Reproduction
of this article is prohibited without written permission from the
American College of Chest Physicians (http://www.chestpubs.org/
site/misc/reprints.xhtml).
DOI: 10.1378/chest.11-2308
e738S

of 0.2 to 0.4 units/mL (Grade 2C). We suggest


that when initiating UFH therapy, UFH boluses
be no greater than 75 to 100 units/kg and that
boluses be withheld or reduced if there are significant bleeding risks (Grade 2C). We suggest
avoiding long-term use of therapeutic UFH in
children (Grade 2C).
1.2. We suggest, for neonates and children receiving either once- or twice-daily therapeutic lowmolecular-weight heparin (LMWH) that the
drug be monitored to a target anti-Xa activity
range of 0.5 to 1.0 units/mL in a sample taken
4 to 6 h after subcutaneous injection or 0.5 to
0.8 units/mL in a sample taken 2 to 6 h after
subcutaneous injection (Grade 2C).
1.3. We suggest, for children receiving vitamin K
antagonists (VKAs), that the drug be monitored
to a target international normalized ratio (INR)
of 2.5 (range, 2.0-3.0), except in the setting of
prosthetic cardiac valves where we suggest adherence to the adult recommendations outlined in
the article by Whitlock et al in this supplement
(Grade 2C). We suggest that INR monitoring
with point-of-care monitors be made available
where resources make this possible (Grade 2C).
1.5. We suggest that when aspirin is used for
antiplatelet therapy in children, it is used in doses
of 1 to 5 mg/kg per day (Grade 2C).
2.1. We suggest that central venous access
devices (CVADs) or umbilical venous catheters
(UVCs) associated with confirmed thrombosis
be removed after 3 to 5 days of therapeutic anticoagulation rather than left in situ (Grade 2C).
We suggest either initial anticoagulation or supportive care with radiologic monitoring for
extension of thrombosis rather than no follow-up
(Grade 2C); however, in previously untreated
patients, we recommend the start of anticoagulation if extension occurs (Grade 2C). We suggest that anticoagulation should be with either
(1) LMWH or (2) UFH followed by LMWH. We
suggest a total duration of anticoagulation of
between 6 weeks and 3 months rather than
shorter or longer durations (Grade 2C). If either
a CVAD or a UVC is still in place on completion
of therapeutic anticoagulation, we suggest a
prophylactic dose of anticoagulation until such
time as the CVAD or UVC is removed (Grade 2C).
We suggest against thrombolytic therapy for
neonatal VTE unless major vessel occlusion
is causing critical compromise of organs or
limbs (Grade 2C). We suggest if thrombolysis is
required, tissue plasminogen activator (tPA)
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

is used rather than other lytic agents (Grade 2C),


and we suggest plasminogen (fresh frozen plasma
[FFP]) administration prior to commencing
therapy (Grade 2C).
2.2. For unilateral renal vein thrombosis (RVT)
in the absence of renal impairment or extension
into the inferior vena cava (IVC), we suggest
either (1) supportive care with radiologic monitoring for extension of thrombosis (if extension
occurs we suggest anticoagulation) or (2) anticoagulation with UFH/LMWH or LMWH in
therapeutic doses rather than no therapy.
If anticoagulation is used, we suggest a total
duration of between 6 weeks and 3 months
rather than shorter or longer durations of therapy (Grade 2C). For unilateral RVT that extends
into the IVC, we suggest anticoagulation with
UFH/LMWH or LMWH for a total duration of
between 6 weeks and 3 months (Grade 2C).
2.3. For bilateral RVT with evidence of renal
impairment, we suggest anticoagulation with
UFH/LMWH or initial thrombolytic therapy
with tPA followed by anticoagulation with UFH/
LMWH (Grade 2C).
2.4. For neonates with CVADs, we recommend
to maintain CVAD patency with UFH continuous
infusion at 0.5 units/kg per h over no prophylaxis (Grade 1A) or intermittent local thrombolysis
(Grade 2C). For neonates with blocked CVADs,
we suggest local thrombolysis after appropriate
clinical assessment (Grade 2C).
2.6. For neonates and children having modified
Blalock-Taussig shunts (MBTS), we suggest intraoperative UFH therapy (Grade 2C). For neonates
and children after MBTS surgery, we suggest
either aspirin or no antithrombotic therapy as compared with prolonged LMWH or VKAs (Grade 2C).
2.9. For neonates and children with acute femoral artery thrombosis, we recommend therapeutic
doses of IV UFH as initial therapy compared
with aspirin or no therapy (Grade 1B) or LMWH
(Grade 2C). We suggest subsequent conversion
to LMWH, or else continuation of UFH, to complete 5 to 7 days of therapeutic anticoagulation
as compared with a shorter or longer duration
(Grade 2C).
2.10. For neonates and children with limbthreatening or organ-threatening (via proximal
extension) femoral artery thrombosis who fail
to respond to initial UFH therapy and who
have no known contraindications, we recommend
thrombolysis (Grade 1C). For neonates and chilwww.chestpubs.org

dren with femoral artery thrombosis, we recommend surgical intervention compared with UFH
therapy alone when there is a contraindication
to thrombolytic therapy and organ or limb death
is imminent (Grade 1C).
2.11. For neonates and children with peripheral
arterial catheters in situ, we recommend UFH
continuous infusion at 0.5 units/mL at 1 mL/h
compared with normal saline (Grade 1A).
2.12. For neonates and children with a peripheral arterial catheter-related TE, we suggest
immediate removal of the catheter (Grade 2B).
For neonates and children with a symptomatic
peripheral arterial catheter-related TE, we
suggest UFH anticoagulation with or without
thrombolysis or surgical thrombectomy and
microvascular repair with subsequent heparin
therapy (Grade 2C).
2.13. For neonates with umbilical arterial catheters (UACs), we suggest UAC placement in a
high rather than a low position (Grade 2B).
2.14. For neonates with UAC, we suggest prophylaxis with a low-dose UFH infusion via the
UAC (heparin concentration of 0.25-1 unit/mL,
total heparin dose of 25-200 units/kg per day) to
maintain patency (Grade 2A).
2.16. For neonates and children requiring
cardiac catheterization (CC) via an artery, we
recommend administration of IV UFH as thromboprophylaxis over no prophylaxis (Grade 1A) or
aspirin (Grade 1B). For neonates and children
requiring CC via an artery, we recommend the
use of UFH doses of 100 units/kg as a bolus compared with a 50-unit/kg bolus (Grade 1B). In prolonged procedures, we suggest further doses of
UFH rather than no further therapy (Grade 2B).
2.17. For neonates with cerebral sinovenous
thrombosis (CSVT) without significant intracranial hemorrhage, we suggest anticoagulation, initially with UFH or LMWH and subsequently with
LMWH, for a total therapy duration between
6 weeks and 3 months rather than shorter or
longer treatment duration (Grade 2C). For neonates with CSVT with significant hemorrhage,
we suggest either (1) anticoagulation or (2) supportive care with radiologic monitoring of the
thrombosis at 5 to 7 days and anticoagulation if
thrombus extension is noted as compared with
no therapy (Grade 2C).
2.18. For neonates with a first arterial ischemic
stroke (AIS), in the absence of a documented,
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e739S

ongoing cardioembolic source, we suggest supportive care over anticoagulation or aspirin


therapy (Grade 2C).

anticoagulant therapy beyond 3 months in either


therapeutic or prophylactic doses until the risk
factor has resolved (Grade 2C).

2.19. For neonates with a first AIS and a documented cardioembolic source, we suggest anticoagulation with UFH or LMWH (Grade 2C).

2.22.4. In children with recurrent idiopathic


VTE, we recommend indefinite treatment with
VKAs (Grade 1A).

2.20. For neonates with recurrent AIS, we suggest anticoagulant or aspirin therapy (Grade 2C).

2.22.5. In children with recurrent secondary


VTEs with an existing reversible risk factor for
thrombosis, we suggest anticoagulation until
resolution of the precipitating factor but for a
minimum of 3 months as compared with no further therapy (Grade 2C).

2.21. For neonates with clinical presentations of


homozygous protein C deficiency, we recommend
administration of either 10 to 20 mL/kg of FFP
every 12 h or protein C concentrate, when available, at 20 to 60 units/kg until the clinical lesions
resolve (Grade 1A). For neonates with homozygous protein C deficiency, after initial stabilization, we recommend long-term treatment with
VKA (Grade 1C), LMWH (Grade 1C), protein C
replacement (Grade 1B), or liver transplantation
(Grade 1C) compared with no therapy.
2.22.1. In children with first VTE (CVAD and
non-CVAD related) we recommend acute anticoagulant therapy with either UFH or LMWH
(Grade 1B). We recommend initial treatment with
UFH or LMWH for at least 5 days (Grade 1B).
For ongoing therapy, we recommend LMWH
or UFH. For patients in whom clinicians will
subsequently prescribe VKAs, we recommend
beginning oral therapy as early as day 1 and discontinuing UFH/LMWH on day 6 or later than
day 6 if the INR has not exceeded 2.0 compared
with no therapy (Grade 1B).
2.22.2. We suggest that children with idiopathic
VTE receive anticoagulant therapy for 6 to
12 months compared with no therapy (Grade 2C).
Underlying values and preferences: Families who
place a high value on avoiding the unknown risk of
recurrence in the absence of an ongoing risk factor
and a lower value on avoiding the inconvenience of
therapy or potential impact of therapy on growth
and development and bleeding risk associated with
antithrombotic therapy are likely to choose to continue anticoagulant therapy beyond 6 to 12 months.
2.22.3. In children with secondary VTE (ie, VTE
that has occurred in association with a clinical
risk factor) in whom the risk factor has resolved,
we suggest anticoagulant therapy be administered for 3 months (Grade 2C) as compared
with no further therapy. In children who have
ongoing, but potentially reversible risk factors,
such as active nephrotic syndrome or ongoing
asparaginase therapy, we suggest continuing
e740S

2.22.6. In children with a CVAD in place who


have a VTE, if a CVAD is no longer required or
is nonfunctioning, we recommend it be removed
(Grade 1B). We suggest at least 3 to 5 days of
anticoagulation therapy prior to its removal
rather than no anticoagulation prior to removal
(Grade 2C). If CVAD access is required and the
CVAD is still functioning, we suggest that the
CVAD remain in situ and the patient be given anticoagulants (Grade 2C). For children with a first
CVAD-related VTE, we suggest initial management
as for secondary VTE as previously described.
2.22.7. In children with CVAD in place who
have a VTE and in whom the CVAD remains
necessary, we suggest, after the initial 3 months
of therapy, that prophylactic doses of VKAs (INR
range, 1.5-1.9) or LMWH (anti-Xa level range,
0.1-0.3 units/mL) be given until the CVAD is
removed (Grade 2C). If recurrent thrombosis
occurs while the patient is receiving prophylactic therapy, we suggest continuing therapeutic
doses until the CVAD is removed and for a minimum of 3 months following the VTE (Grade 2C).
2.23. In children with VTE, we suggest that
thrombolysis therapy be used only for life- or
limb-threatening thrombosis (Grade 2C). If thrombolysis is used in the presence of physiologically
low levels or pathologic deficiencies of plasminogen, we suggest supplementation with plasminogen (Grade 2C). In children with VTE in
whom thrombolysis is used, we suggest systemic
thrombolysis or catheter-directed thrombolysis,
depending on institutional experience and, in
the latter case, technical feasibility.
2.24. In children with life-threatening VTE, we
suggest thrombectomy (Grade 2C). In children
who have had a thrombectomy, we suggest anticoagulant therapy as per recommendation (Recommendation 2.22) (Grade 2C). In children . 10 kg
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

body weight with lower-extremity VTE and a


contraindication to anticoagulation, we suggest
placement of a retrievable IVC filter (Grade 2C).
In children who receive a filter, we suggest that
the filter be removed as soon as possible if
thrombosis is not present in the basket of the
filter and when contraindication to anticoagulation is resolved (Grade 2C). In children who
receive an IVC filter, we recommend appropriate anticoagulation for VTE (see Recommendation 1.2) as soon as the contraindication to
anticoagulation is resolved (Grade 1C).
2.25. In children with cancer, we suggest that
management of VTE follow the general recommendations for management of VTE in children.
We suggest the use of LMWH in the treatment
of VTE for a minimum of 3 months until the precipitating factor has resolved (eg, use of asparaginase) (Grade 2C).
Remarks: The presence of cancer, the need for surgery, chemotherapy, or other treatments may modify
the risk-benefit ratio for treatment of VTE, and clinicians should consider these factors on an individual
basis.
2.26. For children with VTE in the setting of antiphospholipid antibodies (APLAs), we suggest
management as per general recommendations
for VTE management in children.
2.27. For children with VTE, independent of
the presence or absence of inherited thrombophilic risk factors, we suggest that the duration and intensity of anticoagulant therapy as
per Recommendation 2.22.
2.28. For children with first VTE secondary to
structural venous abnormalities, we suggest
anticoagulation as per other spontaneous VTE
(Recommendation 2.22) and consideration of
subsequent percutaneous or surgical interventions, depending on patient factors and institutional experience. For children with recurrent
VTE secondary to structural venous abnormalities, we suggest indefinite anticoagulation unless
successful percutaneous or surgical interventions can be performed (Grade 2C).
2.29. For children with right atrial thrombosis
related to CVAD, we suggest removal of the CVAD
with or without anticoagulation, depending on
the individual risk factors, compared with leaving
the CVAD in situ (Grade 2C). For children with
large (. 2 cm) mobile right atrial thrombosis,
we suggest anticoagulation, with appropriately
www.chestpubs.org

timed CVAD removal, and consideration of surgical intervention or thrombolysis based on


individualized risk-benefit assessment compared
with no anticoagulation therapy (Grade 2C).
2.30. For CVADs, we suggest flushing with normal saline or heparin or intermittent recombinant urokinase (rUK) to maintain patency
as compared with no therapy (Grade 2C). For
blocked CVADs, we suggest tPA or rUK to
restore patency (Grade 2C). If after at least
30 min following local thrombolytic instillation CVAD patency is not restored, we suggest a second dose be administered. If the
CVAD remains blocked following two doses of
local thrombolytic agent, we suggest radiologic
imaging to rule out a CVAD-related thrombosis
(Grade 2C).
2.31. For children with short- or medium-term
CVADs, we recommend against the use of routine systemic thromboprophylaxis (Grade 1B).
2.34. For children receiving long-term home
total parenteral nutrition (TPN), we suggest
thromboprophylaxis with VKAs (Grade 2C).
2.35. For children who have bilateral cavopulmonary shunt (BCPS), we suggest postoperative
UFH (Grade 2C).
2.36. For children after Fontan surgery, we recommend aspirin or therapeutic UFH followed
by VKAs over no therapy (Grade 1C).
2.37. For children having endovascular stents
inserted, we suggest administration of UFH
perioperatively (Grade 2C).
2.38. For pediatric patients with cardiomyopathy,
we suggest VKAs no later than their activation
on a cardiac transplant waiting list (Grade 2C).
Underlying values and preferences: Parents who place a
high value on avoiding the inconvenience, discomfort,
and limitations of anticoagulant monitoring and a
lower value on the uncertain reduction in thrombotic
complications are unlikely to choose VKA therapy for
their children who are eligible for transplant.
2.39. For children with primary pulmonary hypertension, we suggest starting anticoagulation
with VKAs at the same time as other medical
therapy (Grade 2C).
2.40-2.42. For children with biologic or mechanical prosthetic heart valves, we recommend that
clinicians follow the relevant recommendations
from the adult population.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e741S

2.44. For children with ventricular assist devices


(VADs), we suggest administration of UFH
(Grade 2C). We suggest starting UFH between
8 and 48 h following implantation (Grade 2C).
In addition, we suggest antiplatelet therapy
(either aspirin or asprin and dipyridamole) to commence within 72 h of VAD placement (Grade 2C).
For children with VAD, once clinically stable,
we suggest switching from UFH to either LMWH
or VKA (target INR 3.0 range, 2.5-3.5) until
transplanted or weaned from VAD (Grade 2C).
2.45. For patients undergoing hemodialysis via
an arteriovenous fistula, we suggest routine use
of VKAs or LMWH as fistula thromboprophylaxis
as compared with no therapy (Grade 2C).
2.46. For patients undergoing hemodialysis
via CVAD, we suggest routine use of VKAs or
LMWH for thromboprophylaxis as compared
with no therapy (Grade 2C).
2.47. For children having hemodialysis, we suggest the use of UFH or LMWH during hemodialysis to maintain circuit patency independent
of type of vascular access (Grade 2C).
2.48. For children with Kawasaki disease, we
recommend aspirin in high doses (80-100 mg/kg
per day during the acute phase for up to 14 days)
as an antiinflammatory agent, then in lower
doses (1-5 mg/kg per day for 6 to 8 weeks) as an
antiplatelet agent (Grade 1B). For children with
Kawasaki disease, we recommend IV g-globulin
(2 g/kg, single dose) within 10 days of the onset
of symptoms (Grade 1A).
2.49. For children with moderate or giant coronary aneurysms following Kawasaki disease, we
suggest that warfarin in addition to low-dose
aspirin be given as primary thromboprophylaxis
(Grade 2C).

(Grade 2C). For children with CSVT with significant hemorrhage, we suggest initial anticoagulation as for children without hemorrhage or
radiologic monitoring of the thrombosis at 5 to
7 days and anticoagulation if thrombus extension is noted at that time (Grade 2C). In children
with CSVT and potentially recurrent risk factors
(for example, nephrotic syndrome, asparaginase
therapy), we suggest prophylactic anticoagulation at times of risk factor recurrence (Grade 2C).
We suggest thrombolysis, thrombectomy, or
surgical decompression only in children with
severe CSVT in whom there is no improvement
with initial UFH therapy (Grade 2C).
2.52. For children with acute AIS, with or without thrombophilia, we recommend UFH or
LMWH or aspirin as initial therapy until dissection and embolic causes have been excluded
(Grade 1C). For children with acute AIS, we
suggest, once dissection and cardioembolic
causes are excluded, daily aspirin prophylaxis
for a minimum of 2 years as compared with
no antithrombotic therapy (Grade 2C). For children receiving aspirin who have recurrent AIS
or transient ischemic attacks (TIAs), we suggest changing to clopidogrel or anticoagulant
therapy with LMWH or VKA (Grade 2C). For
children with AIS, we recommend against the
use of thrombolysis (tPA) or mechanical thrombectomy outside of specific research protocols
(Grade 1C).
2.53. For AIS secondary to cardioembolic causes,
we suggest anticoagulant therapy with LMWH
or VKAs for at least 3 months (Grade 2C). For
AIS secondary to cardioembolic causes in children with demonstrated right-to-left shunts (eg,
patent foramen ovale [PFO]), we suggest surgical closure of the shunt (Grade 2C).

2.50. For children with Kawasaki disease who


have giant aneurysms and acute coronary artery
thrombosis, we suggest thrombolysis or acute
surgical intervention (Grade 2C).

2.54. For AIS secondary to dissection, we suggest anticoagulant therapy with LMWH or VKAs
for at least 6 weeks (Grade 2C). Ongoing treatment will depend on radiologic assessment of
degree and extent of stenosis and evidence of
recurrent ischemic events.

2.51. For children with CSVT without significant intracranial hemorrhage, we recommend
anticoagulation initially with UFH or LMWH
and subsequently with LMWH or VKA for a
minimum of 3 months relative to no anticoagulation (Grade 1B). In children who after 3 months
of therapy still experience occlusion of CSVT
or ongoing symptoms, we suggest administration of a further 3 months of anticoagulation

2.55. For children with acute AIS secondary to


non-Moyamoya vasculopathy, we recommend
UFH or LMWH or aspirin for 3 months as
initial therapy compared with no treatment
(Grade 1C). For children with AIS secondary
to non-Moyamoya vasculopathy, we suggest ongoing antithrombotic therapy should be guided
by repeat cerebrovascular imaging.

e742S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

2.56. For children with acute AIS secondary to


Moyamoya, we suggest aspirin over no treatment as initial therapy (Grade 2C).
2.57. For children with Moyamoya, we suggest
they be referred to an appropriate center for
consideration of revascularization.
(TE) in pediatric patients is rare
Thromboembolism
and makes management studies a challenge, result-

ing in limited direct evidence. In children, 50% of


all drugs used are unlicensed or off-label, reflecting
the paucity of specific trials in children.1,2 Thus, most
recommendations are based on extrapolation from
adults. There is evidence that such extrapolation may,
in many circumstances, be inappropriate.3-5
Fortunately, recent regulatory initiatives have
resulted in the development of specific pediatric
investigational plans for select novel anticoagulants.6
Although these studies will take years to complete,
they will provide excellent data on the safety and efficacy of currently used anticoagulants in children as
well as an understanding of the newer drugs. At the
same time, additional research is required to understand the basic pharmacokinetics and pharmacodynamics of commonly prescribed antithrombotic drugs
in children because significant differences exist in
antithrombotic activity and impact on monitoring
tests in children compared with adults.4,7
This article is divided into two sections. The first
section details the evidence showing that the interaction of antithrombotic agents with the hemostatic
system of the young differs from that of adults. This
section describes the pediatric-specific aspects of
mechanisms of action; therapeutic ranges; dose regimens; monitoring requirements; factors influencing
dose-response relationships; and side effects of antithrombotic, antiplatelet, and thrombolytic agents. The
second section provides the evidence and recommendations for antithrombotic therapy in specific clinical
situations in neonates and children.
In managing children with antithrombotic therapy,
as with any therapy, the values and preferences of
the patient and family are crucial to consider in the
treatment algorithms. Preliminary studies suggest that
these values and preferences can vary widely among
families, perhaps related to culture and religion, but
certainly reflect the variation in patient and parental
personal views and experiences.8
Throughout this article, the term pediatric patients
refers to all neonates and children (birth-18 years).
Neonates refers to infants from birth to 28 days
corrected for gestational age. Children refers to
patients aged 28 days to 18 years. The age at which
adolescents should be considered adults from the

www.chestpubs.org

perspective of treatment guidelines remains controversial. Young adults (18-25 years) are sparsely represented in most adult data about management of TE.
In other areas of medicine, this demographic is being
recognized as a separate entity, which requires specific study.9 In addition to chronologic age, clinicians
need to consider factors such as physical development, stage of puberty, and emotional and intellectual development. Adolescents are transitioned to
adult services after they leave school or between
16 and 21 years of age, depending on their local jurisdiction. In addition, there is considerable variation
based on individual circumstances.
Comprehensive literature searches were performed
as per the American College of Chest Physicians
(ACCP) guidelines based on the questions presented
in Table 1, and recommendations are based on the
ACCP grades of recommendation.10 Where possible,
because of the physiologic and pathophysiologic differences as well as the markedly different implications of therapy, recommendations are presented for
neonates and children separately. However, in cases
where the available data do not adequately differentiate between the two age groups, the combined recommendations are presented.
1.0 Antithrombotic Therapy
in Pediatric Patients
The use of antithrombotic drugs in pediatric patients
differs from adults.11 First, the epidemiology of TE in
pediatric patients differs from that seen in adults.12-24
Second, the hemostatic system is a dynamic, evolving
entity that likely affects not only the frequency and
natural history of TEs in children but also the response
to therapeutic agents.4,25,26 Third, the distribution,
binding, and clearance of antithrombotic drugs are
age dependent.27-29 Fourth, the frequency and type
of intercurrent illnesses and concurrent medications
vary with age. Fifth, the need for general anesthesia to
perform many diagnostic studies in pediatric patients
has an impact on the ability to investigate and monitor TEs and, hence, the confidence one can have in
therapeutic decisions. Sixth, limited vascular access
reduces the ability to effectively deliver some antithrombotic therapies and can influence the choice of
antithrombotic agent. Often, the only vascular access
available is used for drug delivery, so accurate monitoring of blood anticoagulant levels is difficult. Seventh, specific pediatric formulations of antithrombotic
drugs are not available, making accurate, reproducible dosing difficult, which is especially the case
for vitamin K antagonists (VKAs) (no suspension/liquid preparation) and low-molecular-weight heparin
(LMWH) (in many countries, the most readily available
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e743S

e744S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age) and
children

Neonates (premature
and term up to 28 d
corrected age) and
children

2.1

2.2-2.3

2.2-2.3

2.4-2.5

2.4-2.5

2.6

2.7

Section

Treatment

Prophylaxis

Prophylaxis

Prophylaxis

Treatment

Treatment

Treatment

Informal Question

Blalock-Taussig
shunt-blocked

Blalock-Taussig shunt

CVAD

CVAD

Renal vein thrombosisbilateral or IVC


involvement

Renal vein thrombosisunilateral

DVT (CVL and


non-CVL related), PE

Population

Thrombolysis

Anticoagulation (heparin or
LMWH), aspirin,
clopidogrel

Systemic heparin or LMWH


prophylaxis

Local heparin (1-2 units/mL


infusion) or heparin
lock, intermittent local
thrombolysis

Anticoagulation,
thrombolysis

Anticoagulation

Anticoagulation,
thrombolysis

Intervention (s)

Surgical intervention

No therapy, each other

No therapy, each other

No therapy, each other

No therapy, each other

No therapy

No therapy, each other

Comparator

PICO Question
Outcome
Mortality
Pulmonary embolus
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
Renal failure
Renal atrophy
Hypertension
Extension
Recurrent VTE
Hemorrhage (major and CNS)
Mortality
Renal failure
Renal atrophy
Hypertension
Extension
Recurrent VTE
Hemorrhage (major and CNS)
Patency
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS)
Patency
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS
Intracardiac thrombosis
(includes shunt thrombosis)
Mortality
Tissue loss
Hemorrhage (major and CNS)
Intracardiac thrombosis
(includes shunt thrombosis)
Mortality
Tissue loss
Hemorrhage (major and CNS)

Table 1[Introduction] PICO Questions for Antithrombotic Therapy in Neonates and Children

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

Methodology

www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e745S

Neonates (premature
and term up to 28 d
corrected age) and
children
Neonates (premature
and term up to 28 d
corrected age) and
children
Neonates (premature
and term up to 28 d
corrected age) and
children
Neonates (premature
and term up to 28 d
corrected age) and
children
Neonates (premature
and term up to 28 d
corrected age)
Neonates (premature
and term up to 28 d
corrected age)

2.9-2.10

Neonates (premature
and term up to 28 d
corrected age)

Neonates (premature
and term up to 28 d
corrected age) and
children

2.15

2.16

2.13-2.14

2.13-2.14

2.12

2.11

2.9-2.10

Neonates (premature
and term up to 28 d
corrected age)

2.8

Section

Prophylaxis

Treatment

Prophylaxis

Prophylaxis

Treatment

Prophylaxis

Treatment

Treatment

Prophylaxis

Informal Question

Population

Cardiac catheter

Aortic thrombosis
(UAC related or
spontaneous)

UAC

UAC

Peripheral arterial
thrombosis (excluding
femoral artery)

Peripheral arterial
catheters (excluding
femoral artery)

Femoral artery
thrombosis

Femoral artery
thrombosis

Stage 1 Norwood

Intervention (s)

Heparin prophylaxis,
Aspirin prophylaxis

Thrombolysis

Heparin prophylaxis

Exposure (high
position [. T10])

Thrombolysis

Thrombolysis (followed by
standard anticoagulation
or antiplatelet therapy),
thrombectomy
Thrombolysis

Anticoagulation

Anticoagulation

Outcome

No therapy

Anticoagulation

No therapy

No therapy,
thrombectomy,
anticoagulation,
antiplatelet therapy
Low position (L3-L5)

Aortic thrombosis
NEC
Hemorrhage (major and CNS)
Patency
Aortic thrombosis
Hemorrhage (major and CNS)
NEC
Embolization (eg, digital artery)
Tissue loss
Mortality
Mortality
Tissue loss
Renal impairment
Hypertension
NEC
Embolization
Hemorrhage (major and CNS)
Femoral artery thrombosis
Embolization non-CNS
Cardioembolic stroke
Hemorrhage (major and CNS)

Tissue loss
Growth failure
Hemorrhage (major and CNS)

Intracardiac thrombosis
Mortality
Tissue loss
Hemorrhage (major and CNS)
No therapy or
Claudication
antiplatelet therapy
Leg shortening
Tissue loss
Hemorrhage (major and CNS)
Anticoagulation or
Claudication
antiplatelet therapy
Leg shortening
(without thrombolysis), Tissue loss
each other
Hemorrhage (major and CNS)
No therapy
Tissue loss
Growth failure
Hemorrhage (major and CNS)

Antiplatelet therapy

Comparator

PICO Question

Table 1Continued

Methodology

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

e746S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

Neonates (premature
and term up to 28 d
corrected age)

2.21

Children (day 28 to
16-18 y)

Neonates (premature
and term up to 28 d
corrected age)

2.18-2.20

2.23

Neonates (premature
and term up to 28 d
corrected age)

2.18-2.20

Children (day 28 to
16-18 y)

Neonates (premature
and term up to 28 d
corrected age)

2.18-2.20

2.22

Neonates (premature
and term up to 28 d
corrected age)

2.17

Section

Treatment

Treatment

Treatment

Treatment

Treatment

Treatment

Treatment

Informal Question

Population

DVT (CVL and non-CVL


related), PE

DVT (CVAD and nonCVAD related), PE

Purpura fulminans

AIS (recurrent)

AIS (unknown vs embolic


vs traumatic/dissection
vs thrombophilia) (no
documented ongoing
cardioembolic source)
AIS (documented
cardioembolic source)

CSVT

Intervention (s)

Comparator

No therapy, each other

Anticoagulation

No therapy

Antiplatelet therapy
or no therapy

No therapy

No therapy

Systemic thrombolysis
Anticoagulation
(in conjunction with
anticoagulant therapy),
local thrombolysis
pharmacomechanical
thrombolysis (in conjunction
with anticoagulant therapy)

Anticoagulation

Protein C replacement,
fresh frozen plasma

Antiplatelet therapy or
anticoagulation

Anticoagulation

Anticoagulation

Anticoagulation

PICO Question

Table 1Continued

Outcome

Methodology

Mortality
Functional status
Hemorrhage (major and CNS)
Recurrent AIS (rare)
Mortality
Functional status
Hemorrhage (major and CNS)
Recurrent AIS (rare)
Mortality
Vision
Neurologic outcome
Primary thrombosis
Recurrent thrombosis (among
those with major vessel
thrombosis at presentation)
Mortality
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Phlegmasia cerulea dolens

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

Mortality
RCT, observational
Functional status
studies
Extension
Recurrent CSVT
Hemorrhage (major and CNS)
Visual outcomes/need for surgical
management of increased ICP
(fenestration shunt)
Mortality
RCT, observational
Functional status
studies
Hemorrhage (major and CNS)
Recurrent AIS (rare)

www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e747S

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y) with
cancer or
leukemia

Children (day 28 to
16-18 y) with
antiphospholipid
antibodies or lupus
anticoagulant

Children (day 28
to 16-18 yr)
with positive
thrombophilia

Children (day 28 to
16-18 y) with
structural venous
abnormality

Children (day 28 to
16-18 y)

2.24

2.25

2.26

2.27

2.28

2.29

Section

Treatment

Treatment

Treatment
duration/
intensity

Treatment
duration/
intensity

Treatment
duration/
intensity

Treatment

Informal Question

Right atrial thrombosis


( CVAD related)

DVT (CVL and non-CVL


related), PE

DVT (CVAD and


non CVAD related), PE

DVT (CVL and non-CVL


related), PE

DVT (CVL and non-CVL


related), PE

DVT (CVL and non-CVL


related), PE

Population

Thrombolysis, surgical
thrombectomy
(followed by standard
anticoagulation or
antiplatelet therapy)

Interventional radiology
or surgical stenting,
dilatation or bypass

Anticoagulation,
Interventional radiology
or surgical stenting,
dilatation or bypass

Anticoagulation
(heparin/LMWH)

Anticoagulation
(heparin/LMWH)

Thrombectomy, IVC filter

Intervention (s)

Anticoagulation
(without thrombolysis
or surgical
thrombectomy),
each other

Anticoagulation

No therapy, Each other

VKAs

VKAs

Anticoagulation

Comparator

PICO Question

Table 1Continued

Mortality
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Filter migration or filter fracture
Filter nonretrievability (for
temporary filters)
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Hemorrhage (major and CNS)
Recurrence (DVT or PE)
Mortality
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
Primary Pulmonary embolus
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
Extension
Primary PE
Paradoxical stroke
Postthrombotic syndrome
Recurrence (DVT or PE)
Hemorrhage (major and CNS)
Mortality
PE
Paradoxical stroke
Postthrombotic syndrome
Recurrent VTE
Hemorrhage (major and CNS)

Outcome

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

Methodology

e748S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

Children (day 28 to
16-18 y)

Children (day 28
to 16-18 y)
with positive
thrombophilia

2.30-2.34

2.35

Children (day 28 to
16-18 y)

2.30-2.34

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.30-2.34

2.30-2.34

Prophylaxis

2.30-2.34

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Informal Question

Children (day 28 to
16-18 y)

Section

Population

Glenn or bilateral cavo


pulmonary shunt

CVAD (long term, eg,


home TPN)

CVAD (long term, eg,


oncology)

CVAD (long term, eg,


oncology)

CVAD (short or medium


term, eg, PICU)

CVAD

Intervention (s)

Anticoagulation prophylaxis

Systemic anticoagulation
prophylaxis

Systemic anticoagulation
prophylaxis

Systemic anticoagulation
prophylaxis

Systemic anticoagulation
prophylaxis

Local heparin
(1-2 units/mL infusion),
heparin lock,
intermittent local
thrombolysis

No therapy

No therapy

No therapy

No therapy

No therapy

No therapy

Comparator

PICO Question

Table 1Continued

Outcome
Patency
CVAD dysfunction
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS)
Patency
CVAD dysfunction
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS)
mortality
Patency
CVAD dysfunction
Sepsis/CVAD infection
DVT
PE
Major bleeding
Mortality
Patency
CVAD dysfunction
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS)
Mortality
Postthrombotic syndrome
Patency
CVAD dysfunction
Sepsis/CVAD infection
DVT
PE
Hemorrhage (major and CNS)
Mortality
Postthrombotic syndrome
Intracardiac thrombosis
Mortality
Tissue loss
Hemorrhage (major and CNS)
Ischemic stroke
Fontan surgery

Methodology

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e749S

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.36

2.37

2.38

2.39

2.40-2.42

2.40-2.42

2.40-2.42

2.43

2.44

Section

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Prophylaxis

Informal Question

Population

Ventricular assist
devices

Mechanical prosthetic
heart valves with a
history of thrombotic
events while on
antithrombotic therapy
Bacterial endocarditis

Mechanical prosthetic
heart valves

Biologic prosthetic
heart valves

Primary pulmonary
hypertension

Dilated cardiomyopathy

Endovascular stents

Fontan surgery

Intervention (s)

Anticoagulation,
antiplatelet agents,
prophylaxis

Anticoagulation

Combination,
antiplatelet agents,
and VKAs

Antiplatelet agents,
anticoagulation,
antiplatelet agents,
and VKAs

VKAs or aspirin

VKAs

VKAs or aspirin
prophylaxis

Heparin or LMWH or
aspirin prophylaxis

Anticoagulation,
antiplatelet therapy

No therapy

No therapy

VKAs alone

No therapy

No therapy

No therapy

No therapy

No therapy

No therapy

Comparator

PICO Question

Table 1Continued

Outcome
Intracardiac thrombosis
Mortality
Fontan takedown
Ischemic stroke
Hemorrhage (major and CNS)
Patency
Mortality
Pulmonary emboli
Ischemic stroke
Mortality
Thrombosis
Ischemic stroke
Hemorrhage (major and CNS)
Mortality
Thrombosis
Heart/lung transplantation
Hemorrhage (major and CNS)
Mortality
Valve replacement
Thrombosis
Ischemic stroke
Hemorrhage (major and CNS)
Mortality
Valve replacement
Thrombosis
Ischemic stroke
Hemorrhage (major and CNS)
Mortality
Valve replacement
Thrombosis
Ischemic stroke
Hemorrhage (major and CNS)
Primary embolic stroke
Mortality
Hemorrhage (major and CNS)
Mortality
Thrombosis
Ischemic stroke
Blocked circuit requiring surgery
Hemorrhage (major and CNS)

Methodology

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

e750S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.48-2.50

2.53

Children (day 28 to
16-18 y)

2.48-2.50

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.48-2.50

2.52

Children (day 28
to 16-18 y)

2.47

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.45-2.46

2.51

Prophylaxis

2.45-2.46

Population

Treatment

Treatment

AIS (undetermined
cause, in situ
thrombosis,
thrombophilia)
AIS (cardioembolic)

Kawasaki disease with


coronary aneurysms
and acute coronary
artery thrombosis
CSVT

Treatment

Treatment

Kawasaki disease with


coronary aneurysms

Kawasaki disease

Hemodialysis
(arteriovenous
fistula or CVAD)

Hemodialysis (CVAD)

Hemodialysis
(arteriovenous fistula)

Treatment

Prophylaxis

Prophylaxis
(during
procedure)

Prophylaxis

Informal Question

Children (day 28 to
16-18 y)

Section

Intervention (s)

Anticoagulation

Anticoagulaton,
thrombolysis (followed
by standard
anticoagulation)
Anticoagulation or aspirin,
thrombolysis

Thrombolysis

Anticoagulation

Aspirin, IVIG, aspirin


and IVIG,

Anticoagulation

Continuous
anticoagulation,
procedural UFH
or LMWH

Continuous
anticoagulation,
procedural UFH
or LMWH

Aspirin

No therapy, each other

No therapy, each other

Anticoagulation

Antiplatelet therapy

No therapy

No therapy

No therapy

No therapy

Comparator

PICO Question

Table 1Continued

Outcome

Mortality
Thrombus extension
Functional status
Hemorrhage (major and CNS)
Mortality
Recurrent AIS
Functional status
Hemorrhage (major and CNS)
Mortality
Recurrent AIS
Functional status
Hemorrhage (major and CNS)

Mortality
Thrombosis
Shunt dysfunction
Shunt infection
Hemorrhage (major and CNS)
Mortality
Thrombosis
CVAD dysfunction
Sepsis/CVAD infection
Hemorrhage (major and CNS)
Thrombosis
CVAD dysfunction
Sepsis/CVAD infection
Dialysis failure
Hemorrhage (major and CNS)
Coronary aneurysms
Myocardial infarction
Mortality
Hemorrhage (major and CNS)
Myocardial infarction
Mortality
Hemorrhage (major and CNS)
Myocardial infarction
Mortality
Hemorrhage (major and CNS)

Methodology

(Continued)

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e751S

Prophylaxis

Treatment

Treatment

Treatment

Treatment

Treatment

Informal Question

Population

Sickle cell disease

AIS (sickle cell disease)

AIS (moyamoya,
non-sickle cell)

AIS (moyamoya,
non-sickle cell)

AIS (vasculopathy
other than dissection
or moyamoya)

AIS (dissection)

Intervention (s)

Chronic transfusion
program

Exchange transfusion

Surgical revascularization
(direct/indirect)

Aspirin (with/without
neurosurgical direct/
indirect revascularization),
surgical revascularization
(direct/indirect)

Anticoagulation or aspirin

Anticoagulation

Comparator

No treatment

No antithrombotic
therapy (with/without
neurosurgical direct/
indirect surgical
revascularization),
each other
Antiplatelet therapy
(without direct/
indirect surgical
revascularization)
No treatment

No therapy

Aspirin

Outcome

Mortality
Recurrent AIS
Functional status
Hemorrhage (major and CNS)
Mortality
Recurrent AIS
Functional status
Intracranial hemorrhage
Mortality
Recurrent AIS
Functional status
Intracranial hemorrhage

Mortality
Recurrent AIS
Functional status
Intracranial hemorrhage
Mortality
Recurrent AIS
Functional status
Hemorrhage (major and CNS)
Mortality
Recurrent AIS
Functional status
Hemorrhage (major and CNS)

Methodology

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

RCT, observational
studies

AIS 5 arterial ischemic stroke; CSVT 5 cerebral sinovenous thrombosis; CVAD 5 central venous assist device; CVL 5 central venous line; IVC 5 inferior vena cava; LMWH 5 low-molecular-weight heparin;
NEC 5 necrotizing enterocolitis; PE 5 pulmonary embolus(ism); PICO 5 populations, interventions, comparators, outcomes; PICU 5 pediatric ICU; RCT 5 randomized controlled trial; TPN 5 total
parenteral nutrition; UAC 5 umbilical arterial catheter; UFH 5 unfractionated heparin; VKA 5 vitamin K antagonist.

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.56-2.57

2.58-2.59

Children (day 28 to
16-18 y)

2.56-2.57

Children (day 28 to
16-18 y)

Children (day 28 to
16-18 y)

2.55

2.58-2.59

Children (day 28 to
16-18 y)

2.54

Section

PICO Question

Table 1Continued

predosed syringes are based on adult weights). Eighth,


dietary differences make the use of oral VKAs particularly difficult, which is especially true in neonates
because breast milk and infant formulas have very
different vitamin K levels. Finally, compliance issues
are vastly different, for example, in small infants
who cannot understand the need for therapy, adolescents who intellectually comprehend but emotionally are unable to cooperate, and children who
experience the effects of inadequate parenting. The
social, ethical, and legal implications of these issues frequently interfere with the ability to provide the best
treatment for individual neonates and children.
Recommendation
1.0. We suggest that where possible, pediatric
hematologists with experience in TE manage
pediatric patients with TE (Grade 2C). When this
is not possible, we suggest a combination of a
neonatologist/pediatrician and adult hematologist supported by consultation with an experienced pediatric hematologist (Grade 2C).
1.1 Heparin in Neonates and Children
Unfractionated (or standard) heparin (UFH) is
commonly used in pediatric patients. In tertiary pediatric hospitals, 15% of inpatients are exposed to
UFH each day.30
1.1.1 Mechanism of Action: In this supplement,
Garcia et al31 describe the mechanism of action of
UFH. Table 2 lists the specific factors that may alter
the activities of UFH in children. The clinical implications of these changes on dosing, monitoring,
and effectiveness/safety profile of UFH in children
remains to be fully elucidated.
1.1.2 Therapeutic Range: No clinical outcome
studies have determined the therapeutic range for
UFH in neonates or children. Thus, the therapeutic

range for all indications described in this article is


extrapolated from the therapeutic range for the treatment of VTE in adults. This equates to an activated
partial thromboplastin time (aPTT) that reflects a heparin level by protamine titration of 0.2 to 0.4 units/mL
or an anti-Xa level of 0.35 to 0.7 units/mL.38 The
aPTT therapeutic ranges are universally determined
using adult plasma.
Extrapolating the aPTT range from adults to pediatric patients is unlikely to be valid. For example,
baseline aPTTs in pediatric patients, especially neonates,
often are increased compared with adults. Therefore,
the therapeutic ranges represent a reduced relative
increment in aPTT values.4,26 Schmidt et al39 reported
that routine assays underestimate UFH concentration,
especially in neonates. Recent in vitro and in vivo
data also indicate that the aPTT range that correlates to an anti-Xa level of 0.35 to 0.7 units/mL varies significantly with age and heparin dose.5,27,35-37,40
Further, protamine titration results vary with age,5
and different commercial anti-Xa kits can give substantially different results for anti-Xa measurements
on the same samples probably because of subtle differences in kit formulation. This effect is seen in
both adults and children.35,41,42
These differences are increased in infants in whom
endogenous antithrombin levels are reduced.43 Thus,
the scientific rationale for using a therapeutic range
for UFH in children is increasingly questioned. A
comparative clinical outcome trial comparing monitored therapy (with a target therapeutic range) to
weight-adjusted fixed-dose therapy (unmonitored)
is desperately required. However, in the interim, recommendations must be based on the only published
data available, despite their limitations.
1.1.3 Doses: One prospective cohort study used a
weight-based nomogram to address dosing of UFH
in pediatric patients required to achieve adult therapeutic aPTT values.44 Bolus doses of 75 to 100 units/kg
resulted in therapeutic aPTT values in 90% of children

Table 2[Section 1.1.1] Factors in Children That Affect the Action of UFH
UFH Factor

Age-Related Difference

UFH acts through antithrombin-mediated


catabolism of thrombin and factor Xa

Reduced levels of antithrombin and


prothrombin
Reduced capacity to generate thrombin

UFH is bound to plasma proteins, which


limits free active UFH
Endothelial release of TFPI

Age-related difference in anti-Xa:anti IIa


activity
Alterations in plasma binding

Age-related differences in amount of TFPI


release for same amount of UFH

Evidence
Strong: multiple studies Andrew et al (1990),32
Andrew (1992),33 Andrew et al (1992),34
Monagle et al (2006)26
Strong: multiple studies Ignjatovic et al (2007),35
Andrew et al (1990),32 Andrew (1994)12
Moderate: Newall et al (2010),5 Newall et al
(2009),36 Ignjatovic et al (2006)37
Moderate: Ignjatovic et al (2010),29 Newall et al
(2008),28 Ignjatovic et al (2006),27
Ignjatovic et al (2006)37
Moderate: Ignjatovic et al (2006)37

TFPI 5 tissue factor pathway inhibitor. See Table 1 legend for expansion of other abbreviation.
e752S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

at 4 to 6 h postbolus. Maintenance UFH doses were age


dependent, with infants (up to 2 months old corrected
for gestational age) having the highest requirements
(average, 28 units/kg per h) and children aged . 1 year
having lower requirements (average, 20 units/kg per h).
The doses of UFH required for older children are
similar to the weight-adjusted requirements in adults
(18 units/kg per h).45 Boluses of 75 to 100 units/kg
recently have been shown to result in aPTTs that are
unrecordable for . 100 min.5 This studys implications for the more generalized use of UFH boluses
in children are unknown, but the results suggest that
the recommendations for initial therapy with a bolus
should be reexamined.5
There are few data to support the optimal initial
doses (bolus or infusions) in neonates, especially premature infants. As with all children, consideration of
the individual risk factors for bleeding, and the perceived risk of the thrombosis, will require individualization of the initial dosing strategy until such time
as further studies have been completed.
There are few or no data to define optimal prophylactic doses of UFH. Clinicians commonly use a dose
of 10 units/kg per h as a continuous infusion, although
the efficacy of this has not been proven.46,47
1.1.4 Pharmacokinetics: Studies of UFH in newborns are limited but show that the clearance is faster
than for older children because of a larger volume
of distribution.48,49 Recent studies demonstrate that
following a UFH single bolus in children, the half
life, volume of distribution, clearance, and peak serum
concentration all vary in children compared with adults.
Further, the pharmacokinetics of UFH in children
fits a first-order model and is a function of weight
rather than of age.5
1.1.5 Monitoring: There are now considerable data
showing the lack of correlation among the variety
of monitoring assays available, especially for UFH.
There are no clinical outcome data that recommend

one assay over another or even demonstrate the value


of monitoring.5,35,50 However, given the variability of
dose requirements for UFH to achieve a target range
observed in neonates and children and the increased
clinical risk factors for bleeding, monitoring is the
current standard of care.51,52 Although there are no
published data to support the practice, many clinicians use anti-Xa assays in preference to the aPTT in
children aged , 1 year or for children in pediatric
ICUs (PICUs) because there appears to be a lack of
correlation between the anti-Xa and the aPTT.5,35,50
There are standard dosing nomograms for UFH in
children (Table 3).53
Recommendations for frequency of UFH monitoring are extrapolated from adult practice. The pragmatics of available vascular access, need for painful
procedures, and ability of staff to obtain samples
actually determine the monitoring schedule.3
1.1.6 Adverse Effects: One cohort study reported
major bleeding in one in 65 (1.5%, 95% CI, 0.0%8.3%) children treated with IV UFH for DVT/pulmonary embolism (PE).44 However, many children in
this study were treated with subtherapeutic doses of
UFH. A single-center cohort study reported a major
bleeding rate of 24% (95% CI, 11%-40%) in 38 children who received UFH therapy while in the PICU.54
A common cause of fatal heparin-induced bleeding is accidental overdose, especially in neonates.
The most common cause of this is drug error, with
5,000 units/mL or similar concentration vials being
erroneously selected instead of 50-units/mL vials.
The different uses of heparin in neonatal populations
(from line flushes to extracorporeal circuit support)
usually mean that vastly different-strength heparin
doses are readily available to ward staff. Although
rarely reported in the medical literature, the number
of cases reported in the popular press appears to be
increasing.4,55-60
There are only three case reports of pediatric
UFH-induced osteoporosis.61,62 In two of these, the

Table 3[Section 1.1.5] Protocol for Systemic Heparin Administration and Adjustment for Pediatric Patients
I. Loading dose: heparin 75 units/kg IV over 10 min
II. Initial maintenance dose: 28 units/kg per h for infants aged , 1 y; 20 units/kg per h for children aged . 1 y
III. Adjust heparin to maintain aPTT of 60-85 s (assuming this reflects an anti-Xa level of 0.35-0.70):
aPTT, s

Bolus, units/kg

Hold, min

% Rate Change

50
0
, 50
1 10
50-59
0
0
1 10
60-85
0
0
0
86-95
0
0
210
96-120
0
30
210
0
60
. 120
215
IV. Obtain blood for aPTT 4 h after administration of the heparin loading dose and 4 h after every change in the infusion rate
V. When aPTT values are therapeutic, a daily CBC and aPTT

Repeat aPTT
4h
4h
Next day
4h
4h
4h

Adapted with permission.53 aPTT 5 activated partial thromboplastin time.


www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e753S

patients had additional risk factors before this complication. These limited data, in conjunction with adult
data, would support avoidance of long-term use of
UFH in children when alternative anticoagulants
are available. This recommendation is strengthened
by the physiologic changes in bone seen in childhood,
which potentially places children at increased risk of
osteoporosis compared with adults.63,64
As in adults, the diagnosis of heparin-induced
thrombocytopenia (HIT) in children remains problematic.65 Studies examining the frequency of HIT in
children have varied in their reported results, likely
because of differences in patient inclusion and laboratory techniques.30,66-70 Rates vary from almost zero in
unselected heparinized children30 to 2.3% in children
in the PICU.68 UFH exposure in these cases ranged
from low-dose exposure during heparin flushes used
in maintaining patency of venous access devices
(VADs), to supratherapeutic doses given during cardiopulmonary bypass and hemodialysis. Presumed HIT
currently is the most common indication for children
to receive novel anticoagulant drugs, and the outcome
for these children often is poor because of complications of the novel anticoagulants.71 Danaparoid,
hirudin, and argatroban are alternatives to UFH in
children with HIT.65,72-99
1.1.7 Treatment of Heparin-Induced Bleeding: If
UFH needs to be discontinued for clinical reasons,
termination of the infusion usually will suffice because
of the rapid clearance of UFH. If immediate reversal
is required, protamine sulfate rapidly neutralizes
UFH activity. The required dose of protamine sulfate
is based on the amount of UFH received in the previous 2 h (Table 4).
Recommendation
1.1. We suggest that therapeutic UFH in children
is titrated to achieve a target range of anti-Xa
activity of 0.35 to 0.7 units/mL or an aPTT range
that correlates to this anti-Xa range or to a
protamine titration range of 0.2 to 0.4 units/mL
Table 4[Section 1.1.7] Reversal of Heparin Therapy
Time Since Last
Heparin Dose, min
, 30
30-60
60-120
. 120

Protamine Dose
1.0 mg/100 units heparin received
0.5-0.75 mg/100 units heparin received
0.375-0.5 mg/100 units heparin received
0.25-0.375 mg/100 units heparin received

Maximum dose of 50 mg. Infusion rate of a 10 mg/mL solution should


not exceed 5 mg/min. Hypersensitivity reactions to protamine sulfate
may occur in patients with known hypersensitivity reactions to fish or
those previously exposed to protamine therapy or protamine-containing
insulin.
e754S

(Grade 2C). We suggest that when initiating UFH


therapy, UFH boluses be no greater than 75 to
100 units/kg and that boluses be withheld or
reduced if there are significant bleeding risks
(Grade 2C). We suggest avoiding long-term use
of therapeutic UFH in children (Grade 2C).
1.2 LMWH in Neonates and Children
Despite unproven efficacy, LMWHs have become
the anticoagulant of choice in many pediatric patients
for primary and secondary prophylaxis of TE.100 Potential advantages of LMWH include reduced monitoring
need, lack of interference by other drugs or diet,
reduced HIT, and probable reduced risk of osteoporosis. However, the predictability of the anticoagulant effect with weight-adjusted doses appears to be
reduced compared with adults,101 presumably because
of altered plasma binding.29,102
Throughout the guidelines in this article, we
use the term LMWH and present dosing schedules for a number of different LMWHs. However,
most clinical data with respect to LMWH use in
pediatric patients are derived from studies that used
enoxaparin.52,100,101,103-111
1.2.1 Therapeutic Range: Therapeutic ranges for
LMWH are extrapolated from adults and based on
anti-Xa levels. The guideline for therapeutic LMWHs
being given subcutaneously bid is an anti-Xa level of
0.50 to 1.0 units/mL in a sample taken 4 to 6 h following a subcutaneous injection. The majority of clinical
studies in children have used this therapeutic range
to date, although one study reported using 0.5 to
0.8 units/mL as the target therapeutic range, with
good efficacy and safety outcomes.52 The timing of
testing has varied from 2 to 6 h after dosing.52,108 There
are no comparative studies of alternative therapeutic
ranges in children. For children receiving a dose once
daily, the same target peak level (0.5-0.8 units/mL) or,
alternatively, a trough level of , 0.1 units/mL has
been used.52,108 In children treated with continuous IV
infusions of LMWH, the same target range of 0.5 to
1.0 units/mL has been used.112 The Prophylaxis of
Thromboembolism in Kids Trial (PROTEKT) prophylaxis trial used a target anti-Xa range of 0.1 to
0.3 units/mL measured 4 to 6 h postdose.113
1.2.2 Pharmacokinetics: A two-compartment model
describes enoxaparin kinetics. Body weight is the
most predictive covariate for clearance and central
volume of distribution (clearance, 15 mL/h per kg;
central volume of distribution, 169 mL/kg; intercompartmental clearance, 58 mL/h; peripheral volume
of distribution, 10 L; absorption rate, 0.414/h). Interindividual variability was found to be 54% for clearance and 42% for volume of distribution.52
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

1.2.3 Doses: The subcutaneous doses of LMWH


required in pediatric patients to achieve adult
therapeutic peak anti-Xa levels have been assessed
for enoxaparin, reviparin, dalteparin, and tinzaparin
(Table 5).114-118 Doses have also been reported for
nadroparin.119 Once-daily dosing for enoxaparin has
been described as well.110 (Schobess et al) Although
the initial doses reported in these studies most likely
attain the therapeutic range, considerable interpatient dose differences have been reported, suggesting
the possible need for routine monitoring of anti-Xa
levels in children and neonates.51 This has been
confirmed by recent pharmacokinetic studies of
enoxaparin.52 Initial reports suggested that neonates
require higher-per-kilogram doses than older children, and biologic explanations were proposed.120,121
However, recent studies of nadroparin122 and enoxaparin102 demonstrated a difference in dose requirements across a number of different age groups in
clinical cohort studies, suggesting that some of these
variables may continue to affect drug metabolism
well outside the newborn period. Whether clinical
effectiveness will be altered by having multiple agerelated initial and maintenance dose recommendations is unclear.
Reviews of clinical reports of final dose requirements for neonates have suggested that an even higher
initial dose schedule is more appropriate,123 especially
in preterm infants.111,124,125 However, clinical trials have
not been performed to confirm the safety and efficacy of this approach, and it is difficult to recommend
the optimal initial dosing strategy for term and premature neonates at this time.
IV dosing for enoxaparin has been reported in
one neonate: enoxaparin at 1 mg/kg every 8 h was
required to maintain therapeutic levels.106 Contin-

uous infusion of LMWH (predominantly dalteparin


and nadroparin) has also been described in children.112 Table 5 presents the doses required for
prophylactic LMWH for enoxaparin, reviparin, and
dalteparin.114,115,117
1.2.4 Adverse Events: A recent review of enoxaparin
in neonates (n 5 240 derived from eight articles, four
abstracts, and one review from 1996-2007) reported
that minor side effects were common; major bleeding
was recorded in 13 of 240 (5%) neonates.123 Whether
premature infants are at increased risk is unclear. No
major bleeds were reported in a series of 10 premature neonates.125
A review reported that in 308 children treated with
therapeutic LMWH for venous thrombosis (from
six studies), nine (2.9%) had major bleeding, and
72 (23.4%) had minor bleeding.108 However, at least
one of these studies included neonates.100 The same
review reported that of 133 children treated with prophylactic doses of LMWH for primary prevention of
venous thrombosis, one (0.8%) had major bleeding,
and four (3.0%) had minor bleeding.108 There are no
data addressing the frequency of osteoporosis, HIT,
or other hypersensitivity reactions in children exposed
to LMWH.
Treatment of LMWH-Induced BleedingEquimolar concentrations of protamine sulfate neutralize
anti-IIa activity of LMWH but result in only partial
neutralization of its anti-Xa activity.126 However, in
animal models, LMWH-associated bleeding is completely reversed by protamine sulfate.127-130 The dose of
protamine sulfate required depends on the dose and
type of LMWH used. Repeat doses of protamine may
be required after subcutaneous LMWH. Protocols
for reversal have been published.128

Table 5[Section 1.2.3] Doses of LMWH Used in Pediatric Patients


Drug

Weight

Age

Weight-dependent dose
of reviparin

, 5 kg

na

Age-dependent dose
of enoxaparina

Initial Treatment Dose

Initial Prophylactic Dose

150 u/kg/dose q12h

50 u/kg/dose q12h

. 5 kg

na

100 u/kg/dose q12h

30 u/kg/dose q12h

na

, 2 mo

1.5 mg/kg/dose q12h

0.75 mg//kg/dose q12h

1.0 mg/kg/dose q12h

0.5 mg/kg/dose q12h

129 43 u/kg/dose q24h

92 52 u/kg/dose q24h

na

. 2 mo

Pediatric (all ages) dose


of dalteparinb

na

all

Age-dependent dose
of tinzaparin

na

0-2 mo

275 u/kg

na
na
na
na

2-12 mo
1-5 y
5-10 y
10-16 y

250 u/kg
240 u/kg
200 u/kg
175 u/kg

na 5 not applicable.
a Enoxaparin has 110 anti-factor Xa units/mg.115
b Dalteparin has 100 anti-factor Xa units/mg.
www.chestpubs.org

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e755S

Recommendation
1.2. We suggest, for neonates and children
receiving either once- or twice-daily therapeutic
LMWH, that the drug be monitored to a target
anti-Xa activity range of 0.5 to 1.0 units/mL in a
sample taken 4 to 6 h after subcutaneous injection or 0.5 to 0.8 units/mL in a sample taken 2 to
6 h after subcutaneous injection (Grade 2C).
1.3 VKAs in Neonates and Children
VKAs are problematic in newborns for several
reasons. First, the plasma levels of the vitamin Kdependent coagulation factors are physiologically
decreased in newborns to levels that are comparable to those achieved in adults receiving therapeutic
amounts of VKAs with target international normalized ratios (INRs) of 2.0 to 3.0. Second, infant formula
is supplemented with vitamin K to prevent hemorrhagic disease of the newborn. In contrast, breast
milk has low concentrations of vitamin K, making
breast-fed infants very sensitive to VKAs.131,132 The
latter can be compensated for by feeding breast-fed
neonates 30 to 60 mL of formula each day. Third,
VKAs are only available in tablet form in most countries. Although the tablets can be dissolved in water
for administration to newborns, neither stability data
nor critical assessment of this practice are available.
Fourth, VKAs require frequent monitoring in newborns
because of the rapidly changing physiologic values of
the vitamin K-dependent coagulation proteins and
frequent changes in medications and diet.133 Finally,
although there is substantial information on the use
of VKAs in children aged . 3 months, there is little
efficacy or safety information specific to their use
in neonates. Problems of vascular access, frequent
intercurrent infections, concurrent medications, lack
of liquid preparation, and poor compliance continue
to make VKAs difficult to manage in older children as
well.133
Warfarin is the most commonly used VKA in children.134-138 Acenocoumarol is used in some European
and South American countries.139,140 Phenprocoumon
is the preferred VKA in some parts of Europe. No
data about doses of these other agents have been
reported. At present, the choice of VKA often is influenced by previous experience and familiarity within
a country or region. The remainder of this section
will refer to warfarin unless stated otherwise.
The current therapeutic INR ranges for children
are directly extrapolated from recommendations for
adult patients because there are no clinical trials that
have assessed the optimal INR range for children.
Thus, for most indications, the therapeutic target INR
is 2.5 (range, 2.0-3.0), and the low-dose prophylactic
e756S

target INR is 1.7 (range, 1.5-1.9). Therapeutic ranges


for prosthetic valves are also directly extrapolated
from adult data.
The capacity of plasma from children receiving VKAs
to generate thrombin is delayed and decreased by
25% compared with plasma from adults with similar
INRs,15 suggesting that the INR therapeutic range
for children may be lower than for adults. This hypothesis is further supported by the observation that
plasma concentrations of a marker of endogenous
thrombin generation, prothrombin fragment 1.2, is
significantly lower in children than in adults at similar
INR values.15
1.3.2 Dose Response: An initial dose of 0.2 mg/kg,
with subsequent dose adjustments made according to
an INR nomogram, was evaluated in a prospective
cohort study of children aged , 1 year to 18 years
(Table 6).134 The largest cohort study (n 5 319) found
that infants required an average of 0.33 mg/kg and
teenagers 0.09 mg/kg warfarin to maintain an INR of
2.0 to 3.0.138
1.3.3 Monitoring: Monitoring oral anticoagulant
therapy in children is difficult and requires close supervision with frequent dose adjustments.134,138,141
Point-of-Care Monitoring in Neonates and
ChildrenStudies in children comparing point-of-care
monitors to venipuncture INRs, including comparison
with World Health Organization reference thromboplastins in the laboratory method, have confirmed their
accuracy and reliability.142,143 Although not assessed in
a formalized way, the major advantages identified by
families included reduced trauma of venipunctures,
minimal interruption of school and work, ease of
operation, and portability.143-149
Table 6[Section 1.3.2] Protocol for Oral
Anticoagulation Therapy To Maintain an INR Between
2 and 3 for Pediatric Patients
I
II

III

Day 1: if the baseline INR is 1.0 to 1.3:


Dose 5 0.2 mg/kg orally
Loading days 2-4: if the INR is:
INR
Action
1.1-1.3
Repeat initial loading dose
1.4-1.9
50% of initial loading dose
2.0-3.0
50% of initial loading dose
3.1-3.5
25% of loading dose
. 3.5
Hold until INR , 3.5 then restart at
50% decreased dose
Maintenance oral anticoagulation dose guidelines:
1.1-1.4
Increase by 20% of dose
1.15-1.9
Increase by 10% of dose
2.0-3.0
No change
3.1-3.5
Decrease by 10% of dose
. 3.5
Hold until INR , 3.5, then restart at
20% decreased dose

Adapted with permission.53 INR 5 international normalized ratio.


Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

1.3.4 Adverse Effects of VKAs: Bleeding is the main


complication of VKA therapy. The risk of serious
bleeding in children receiving VKAs for mechanical
prosthetic valves, as calculated across 13 case series,
is , 3.2% per patient-year.150 In one large cohort (comprising 391 patient-years with variable target INR
ranges), the major bleeding rate was 0.5% per patientyear.144 In a randomized trial (n 5 41; target INR
range, 2.0-3.0 for 3 months), major bleeding occurred
in 12.2% (95% CI, 4.1%-26.2%).151 A single-center
study with a nurse-coordinated anticoagulant service
reported major bleeding rates of 0.05% per patientyear.152 Adequate patient and family education protocols have been reported to be a major factor in
reducing adverse bleeding events in children on warfarin therapy.153,154 Nonhemorrhagic complications
of VKAs, such as tracheal calcification or hair loss,
have been described on rare occasions in young children.155,156 Two cohort studies described reduced bone
density in children receiving warfarin for . 1 year.
However, these were uncontrolled studies, and the
role of the underlying disorders in reducing bone density remains unclear.157,158
1.3.5 Treatment of VKA-Induced Bleeding: In the
presence of an excessively prolonged INR (usually . 8)
and no significant bleeding, vitamin K may be used
to reverse the effects of excess anticoagulation. Only
limited data are available in children, but IV vitamin K
in doses of 30 mg/kg have been used.159 Algorithms
for cessation and reversal of warfarin vs cessation
of warfarin alone have not been specifically tested
in children and are extrapolated from adult studies.
In the presence of significant bleeding, immediate
reversal using fresh frozen plasma (FFP) or prothrombin complex concentrates or recombinant factor VIIa
may be required.
1.3.6 Alternative Thrombin Inhibitors: A number
of reports have documented pediatric use of argatroban,79,81,84,86,160,161 bivalirudin,78,162 lepirudin,82,83,87,89,
93,163-165 dabigatran,166,167 danaparoid,75,77,92,96,168-175 and
fondaparinux.161,176-180 Most commonly, these agents
have been used in children with HIT. Two reviews
summarized the clinical indications, doses used, monitoring schedules, and clinical outcomes.71,181
Recommendation
1.3. We suggest, for children receiving VKAs,
that the drug be monitored to a target INR
of 2.5 (range, 2.0-3.0), except in the setting
of prosthetic cardiac valves where we suggest
adherence to the adult recommendations outlined in the article by Whitlock et al in this
supplement (Grade 2C). We suggest that INR
monitoring with point-of-care monitors be made
www.chestpubs.org

available where resources make this possible


(Grade 2C).
1.4 Antiplatelet Drugs in Neonates and Children
1.4.1 Background: Compared with adult controls,
neonatal platelets are hyporeactive to thrombin, adenosine diphosphate/epinephrine, and thromboxane A2.182,183
This hyporeactivity of neonatal platelets is the result
of a defect intrinsic to neonatal platelets.182,183 Paradoxically, the bleeding time is short in newborns because
of increased RBC size, high hematocrit levels, and
increased levels of multimeric forms of von Willebrand factor.184-186 The bleeding time was prolonged,
relative to adults, throughout childhood in two of
three studies.34,187,188
Using the platelet function analyzer (PFA)-100
(Dade International, Inc), cord blood samples from
term neonates were found to have shorter closure
times than samples from older children or adults.189-191
The shorter closure time correlates with the higher
hematocrit levels and increased von Willebrand factor
activity (measured by ristocetin cofactor assay) in
cord blood.190 The multiplate whole-blood aggregometry analyzer also demonstrates age-related differences in platelet reactivity.192 A review has summarized
antiplatelet therapy in children.193
1.5 Aspirin
1.5.1 Therapeutic Range, Dose Response, and
Monitoring: Aspirin remains the most common antiplatelet agent used in pediatrics. The dose of aspirin
for optimal inhibition of platelet aggregation is not
known. Empirical doses of 1 to 5 mg/kg per day have
been proposed.193 Pediatric doses of aspirin are not
based on studies of the effect on platelet function
in pediatric patients.194 The PFA-100 sometimes is
used to monitor aspirin therapy in pediatric patients,
although no data support improved patient outcomes
from this practice.150 The VerifyNow aspirin assay
(Accumetrics, Inc) is a point-of-care device that has
been used to monitor aspirin therapy in adults, but its
use for monitoring aspirin in children has not been
reported.195 Neither the PFA-100 nor the VerifyNow
can be recommended for monitoring aspirin therapy
in children at this time.
1.5.2 Adverse Effects: Neonates may be exposed to
aspirin because of maternal ingestion (eg, treatment
of preeclampsia). Clearance of aspirin is slower in
neonates, potentially placing them at risk for bleeding
for longer periods of time. However, in vitro studies
have not demonstrated an additive effect of aspirin
on platelet hypofunction in newborns, and evidence
linking maternal aspirin ingestion to bleeding in newborns is weak.196 In neonates, additive antiplatelet
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e757S

effect must be considered if concurrent indomethacin therapy is required.


In older children, aspirin rarely causes important
hemorrhage, except in the presence of an underlying
hemostatic defect or in children also treated with
anticoagulants or thrombolytic therapy. The relatively low doses of aspirin used as antiplatelet therapy, compared with the much higher doses used for
antiinflammatory therapy, seldom cause other side
effects. For example, although aspirin is associated
with Reye syndrome, this appears to be a dosedependent effect of aspirin and usually is associated
with doses . 40 mg/kg.197-202
Recommendation
1.5. We suggest that when aspirin is used for
antiplatelet therapy in children, it is used in
doses of 1 to 5 mg/kg per day (Grade 2C).
1.6 Dipyrimadole
Dipyridamole frequently is used as a second-line
antiplatelet agent or in combination with aspirin therapy. Doses of 2 to 5 mg/kg per day are common.203-205
Little in the literature is available on the use of dipyrimadole in children.

1.7.3 Other Antiplatelet Agents: Ticlopidine, another


thienopyridine, is given in doses of 10 mg/kg per day
po q12h (maximum, 250 mg/dose). However, no data
support the use of this drug in children.
The clinically available glycoprotein IIb/IIIa antagonists include IV abciximab, eptifibatide, and tirofiban.210 In one study, children with Kawasaki disease
who were treated with abciximab in addition to standard therapy demonstrated greater regression in
coronary aneurysm diameter at early follow-up than
patients who received standard therapy alone.211
This study compared abciximab to historical controls,
and all patients received additional anticoagulant
therapy.
Treatment of Bleeding Due to Antiplatelet Agents
Antiplatelet agents alone rarely cause serious bleeding in children. More frequently, antiplatelet agents
are one of several other causes of bleeding, such as
an underlying coagulopathy and other antithrombotic
agents. Transfusions of platelet concentrates and
the use of products that enhance platelet adhesion
(plasma products containing high concentrations of
von Willebrand factor or d-des amino arginine vasopressin) may be helpful.

1.7 Clopidogrel

1.8 Thrombolysis in Neonates and Children

1.7.1 Therapeutic Range, Dose Response, and


Monitoring: Clopidogrel is being used with increasing
frequency in children. Initial anecdotal use reported
a dose of 1 mg/kg per day to be effective and safe.
Dosing strategies involved rounding of doses to onequarter or one-half tablets (75-mg tablets). Regular
monitoring of liver and renal function was recommended. The Platelet Aggregation Inhibition on Children on Clopidogrel (PICOLO) study reported that
clopidogrel 0.20 mg/kg per day in children aged birth
to 24 months with cardiac disease, 80% of whom
were also receiving aspirin at a mean dose of 9 mg/kg
per day, achieved a platelet inhibition level (as measured by percent inhibition of adenosine diphosphateinduced platelet aggregation) similar to that which
was targeted in adult studies using 75 mg/d.206 No
other studies have reported laboratory monitoring
of clopidogrel in children.

1.8.1 Background: At birth, plasma concentrations of plasminogen are 50% of adult values
(21 mg/100 mL).212-214 The decreased levels of plasminogen in newborns slows the generation of plasmin215
and reduces the thrombolytic effects of streptokinase,
urokinase and tissue plasminogen activator (tPA)
in an in vitro fibrin clot system.34 A similar response
occurs in children with acquired plasminogen deficiency. Supplementation with plasminogen increases
the thrombolytic effect of all three agents34,216
In pediatric patients, tPA is the agent of
choice.34,216-220 Reasons for this preference include a
previous US Food and Drug Administration warning regarding urokinase, experimental evidence of
improved clot lysis in vitro compared with urokinase
and streptokinase, fibrin specificity, and low immunogenicity.221,222 However, tPA is considerably more
expensive than either streptokinase or urokinase, and
the increased in vitro clot lysis by tPA has not been
demonstrated in clinical trials in children. There is
minimal experience with other thrombolytic agents
in children.217,219,223 A survey showed no consensus in
indications for thrombolysis, dose, mode of delivery,
or duration of therapy.220
Success rates for thrombolysis in pediatric patients
vary. Albisetti217 described complete resolution in
64% of 413 children receiving streptokinase, urokinase, or tPA (53%, 43%, and 69%, respectively).

1.7.2 Adverse Effects: Clinically significant bleeding


episodes were infrequent in the PICOLO trial. High
rates of excessive skin bruising have been reported
when clopidogrel is used in combination with aspirin,
and major bleeding is reported in children receiving
concomitant warfarin therapy.207 Other studies have
reported lower rates, but all studies to date have
been small and retrospective.208 Overdose has been
reported with minimal adverse effects.209
e758S

Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

Twenty-one percent of children had no response to


thrombolytics. Nowak-Gttl et al,218 in a review of
thrombolysis in neonates, reported overall thrombolytic patency rates ranging from 39% for children
with aortic thrombosis to 86.5% for children with cardiac diseases. The collected data suggested no difference in efficacy among streptokinase, urokinase, and
tPA. Differences in success rates were also reported
for thrombolysis in venous vs arterial thrombosis in
children.224 Most studies that reported success or failure of thrombolysis used thrombus resolution as an
outcome measure, with methods of detection ranging from clinical assessment to radiologic assessment.
One series of nine patients used the presence or
absence of postthrombotic syndrome (PTS) as an outcome measure.225 The likelihood of positive reporting
bias and the lack of control groups of patients who
did not receive thrombolytics make the interpretation of these data very difficult.219
1.8.2 Contraindications: Manco-Johnson et al226
described specific contraindications for children, which
included prematurity (, 32 weeks gestation). However, thrombolytics have been successfully given to
increasingly premature babies227-232; the number of
such patients remains small. Clinicians should, in
each case, make an individual assessment of the riskbenefit ratio of thrombolysis.219
1.8.3 Therapeutic Range and Monitoring of
Thrombolytic Agents: There is no therapeutic range
for thrombolytic agents. The correlation between
hemostatic variables and efficacy/safety of thrombolytic therapy is too weak to have useful clinical predictive value.233 However, in patients with bleeding,
the choice and doses of blood products can be guided
by appropriate hemostatic monitoring. The single
most useful assay is fibrinogen level, which usually
can be obtained rapidly and helps to determine the
need for cryoprecipitate or plasma replacement.
A commonly used lower limit for fibrinogen level is
1.0 g/L. The aPTT may not be helpful in the presence
of low fibrinogen levels, concurrent UFH therapy,
and presence of fibrin/fibrinogen degradation products.233 Measurement of fibrin degradation products
or D-dimers are helpful in determining whether a
fibrinolytic effect is present. Maintaining a platelet
count . 100 3 109 during thrombolysis has also been
recommended.226
1.8.4 Doses: Eight case series have reported using
thrombolysis for treatment of non-CNS venous and
arterial thrombosis in children with consistent regimes.
Variable success rates are reported222,224,229,234-238 and
have been summarized recently.219 The most common
dose and duration of tPA was 0.5 mg/kg per h infused
for 6 h.222,224
www.chestpubs.org

Thrombolytic agents are used in low doses to


restore catheter patency. Once again, tPA is the most
commonly used agent; however, use of urokinase
and recombinant urokinase (rUK) have also been
reported.239-248
1.8.5 Route of Administration: No published
studies have compared local to systemic thrombolytic
therapy in children.249 At this time, there is no evidence to suggest an advantage of local over systemic
thrombolytic therapy in children with thrombotic
complications.217 In addition, the small vessel size in
children may increase the risk of local vessel injury
during catheter-directed therapy. The theoretical
advantages of catheter-directed thrombolysis include
the ability to deliver low doses of thrombolytic agent
directly into the thrombus.219 Local therapy may be
appropriate for catheter-related TE when the catheter is already in situ. There are more-recent small
case series reporting catheter-directed thrombolysis
in children.225,250,251
1.8.6 Concurrent Heparin Therapy: Manco-Johnson
et al236 described the use of systemic urokinase
infusions together with low-dose heparin infusions
(10 units/kg per h) for 48 h followed by therapeutic
heparinization and warfarinization in children presenting with a first episode of DVT. Concurrent low-dose
heparin followed by therapeutic heparinization has since
been described in other single-center studies.224,225,229,237
Although concurrent LMWH has also been reported,237
given the bleeding risks of thrombolysis, easily reversible anticoagulation seems more appropriate.
1.8.7 Adverse Effects of Thrombolytic Therapy:
Thrombolytic therapy has significant bleeding complications in children. Early literature reviews (including 255 patients) reported an incidence of bleeding
requiring treatment with packed RBCs of 20%.216
The most frequent problem was bleeding at sites of
invasive procedures. A large single-institution study
reported bleeding in 68% of patients, with bleeding
requiring transfusion occurring in 39%.222 Prolonged
duration of thrombolytic infusion was associated with
increased bleeding.
Zenz et al,238 reported bleeding requiring transfusion in three of 17 (18%) patients treated for between
4 and 11 h and minor bleeding in another nine (54%).
Another recent prospective study reported bleeding
requiring transfusion in three of 26 (11.5%) patients
and minor bleeding episodes in 11 (42%).224 In another
review, Zenz et al232 reported intracerebral hemorrhage (ICH) in 14 of 929 (1.5%) patients. When subdivided according to age, ICH was identified in two
of 468 (0.4%) children after the neonatal period, one
of 83 (1.2%; 95% CI, 0.3%-6.5%) term infants, and
11 of 86 (13.8%; 95% CI, 6.6%-21.7%) preterm
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e759S

infants. However, in the largest study of premature


infants included in this review, the incidence of ICH
was the same in the control arm that did not receive
thrombolytic therapy. A retrospective analysis of
16 newborns who received tPA reported one death
from bleeding.234 Albisetti217 reviewed the literature
and calculated overall incidences (with any thrombolytic agent) of minor and major hemorrhage as
22% and 15%, respectively, with the use of tPA being
associated with incidences of 26% minor and 17%
major hemorrhage.
1.8.8 Treatment of Bleeding Due to Thrombolytic
Therapy: Before thrombolytic therapy is used, clinicians should correct other concurrent hemostatic
problems, such as thrombocytopenia or vitamin K
deficiency. Clinically mild bleeding (eg, oozing from
a wound or puncture site) can be treated with local
pressure and supportive care. Major bleeding may
be treated by stopping the infusion of thrombolytic
agent; administering cryoprecipitate (usual dose of
1 unit/5 kg or 5-10 mL/kg), an antifibrinolytic, or both;
and administering other blood products as indicated.
1.9 Vena Caval Interruption
There are case reports and series reporting the use
of inferior vena cava (IVC) filters in children as young
as 6 years of age.252-257 Increasingly, temporary filters
are used so that they can be retrieved once the risk of
PE is reduced or the contraindication to anticoagulation has resolved. Temporary filters have been left in
situ for . 140 days.258 Raffini et al259 reported an institutional program that resulted in 5% of children presenting with DVT having IVC filters inserted with
minimal adverse outcomes. Chaudry et al260 reported
the use of filters in the internal jugular veins as well as
in the IVC. There are no specific guidelines for the
use of filters in children and the risk-benefit ratio
needs to be considered individually in each case. The
risks will depend in part on patient factors and in part
on the institutional expertise in placing and retrieving
filters.
1.10 Surgical Therapy
Surgical thrombectomy, rarely used in children,
is restricted to situations such as IVC thrombosis in
association with intravascular extension of Wilm tumor,
acute thrombosis of Blalock-Taussig shunts, lifethreatening intracardiac thrombosis immediately
after complex cardiac surgery, prosthetic valve thrombosis, septic thrombosis, and peripheral arterial thrombosis secondary to vascular access in neonates.261-273
There are no specific guidelines for the use of thrombectomy in children, but there is general consensus
that the TE recurrence rate and risk of long-term vase760S

cular damage is high. Clinicians should consider the


risk-benefit ratio individually in each patient, and all
patients should be strongly considered for anticoagulation after the procedure.

2.0 Recommendations for Antithrombotic


Therapy in Specific Clinical Situations
2.1 VTE in Neonates
Two prospective registries from Canada and
Germany collected data on neonatal venous thrombosis, whereas a study from The Netherlands included
data from both neonates and older children.20-22 The
Canadian registry reported mortality according to
the site of the thrombosis; deaths were most frequent (33%) in patients with right atrial/superior
vena caval involvement, but it was not clear how
many of these events were directly attributable to
thrombosis.21 In the German registry, there was one
death due to right atrial/superior vena caval thrombosis, and in the Dutch study, there were no deaths
directly attributable to the presence of thrombosis.20,22
Morbidity following these events is very poorly characterized but includes the development of PTS.
Specific complications such as chylothorax may also
occur depending on the site of the thrombosis. Portal hypertension, which may lead to splenomegaly and
gastroesophageal varices, may occur after umbilical
venous catheter (UVC) thrombosis.274,275 Because
the majority of sick neonates have patent foramen
ovale (PFO), paradoxical emboli causing stroke are
described.276
The incidence of recurrent VTE, PTS, or other
more-specific complications is unknown in treated or
untreated neonates. The following recommendations
are necessarily based on extrapolation of principles
of therapy from adult guidelines, limited clinical
information from registries, individual case studies,
and knowledge of current common clinical practice.
They incorporate the principles of use of anticoagulants and thrombolytics in children as described earlier
in this article. Options for treatment include supportive care only, anticoagulant therapy with either
UFH or LMWH, thrombolytic therapy, and surgery.
Important issues when considering treatment
options in this age group include the site, extent, and
clinical consequences of the thrombosis and the risks
of bleeding complications associated with the use of
anticoagulant or thrombolytic therapy. The latter will
vary considerably with gestational age, birth weight,
and comorbidities, such as lung disease, necrotizing
enterocolitis (NEC), sepsis, and intraventricular hemorrhage (IVH). Management should be individualized
with appropriate consideration of the risk-benefit ratio
for each case. Given the particular risk of paradoxical
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

emboli at the time of central venous access device


(CVAD) removal in neonates with CVAD-related
VTE,276 many clinicians advocate delay in CVAD
removal until 3 to 5 days of anticoagulant therapy
have been given.
As described previously, the majority of studies
for anticoagulation in neonates have been part of
larger studies reporting on children in general and
report use of twice-daily enoxaparin targeted to an
anti-Xa range (measured 4-6 h after dose) of 0.5 to
1.0 units/mL.100,101,103-111 One study reported oncedaily and bid enoxaparin targeted to a range of 0.5 to
0.8 units/mL measured 2 h after dose.52 The relationship between the target therapeutic range and the
clinical outcomes is unknown in neonates.111,123-125
Recommendation
2.1. We suggest that CVADs or UVCs associated
with confirmed thrombosis be removed after
3 to 5 days of therapeutic anticoagulation rather
than left in situ (Grade 2C). We suggest either
initial anticoagulation or supportive care with
radiologic monitoring for extension of thrombosis rather than no follow-up (Grade 2C); however,
in previously untreated patients, we recommend
the start of anticoagulation if extension occurs
(Grade 2C). We suggest that anticoagulation
should be with either (1) LMWH or (2) UFH
followed by LMWH. We suggest a total duration of anticoagulation of between 6 weeks and
3 months rather than shorter or longer durations (Grade 2C). If either a CVAD or a UVC is
still in place on completion of therapeutic anticoagulation, we suggest a prophylactic dose of
anticoagulation until such time as the CVAD or
UVC is removed (Grade 2C). We suggest against
thrombolytic therapy for neonatal VTE unless
major vessel occlusion is causing critical compromise of organs or limbs (Grade 2C). We suggest if thrombolysis is required, tPA is used
rather than other lytic agents (Grade 2C), and
we suggest plasminogen (FFP) administration
prior to commencing therapy (Grade 2C).
2.2-2.3 Renal Vein Thrombosis in Neonates
Renal vein thrombosis (RVT) in neonates is the
most common type of spontaneous venous thrombosis.14,20,21,277 Approximately 25% of cases are bilateral
and 52% to 60% extend into the IVC.14,278 Overall survival now approaches 100%.278-282 Clinical sequelae
in survivors include chronic renal impairment and
hypertension.278-283 Lau et al279 reviewed all Englishlanguage reports of neonatal RVT from 1992 to 2006.
There was no difference in renal outcomes irrespecwww.chestpubs.org

tive of whether the infant received no therapy, UFH,


or LMWH, with 70% of affected kidneys having
irreversible renal atrophy.279 Approximately 20% of
children had hypertension on long-term follow-up,
and 3% of children developed chronic renal failure.
Recurrent thrombosis rates appear low. Thrombophilic
abnormalities did not predict recurrence or outcome;
hence, their presence or absence are not useful in
determining initial therapy. Because the existing direct
evidence suggests no benefit, anticoagulant and thrombolytic therapy remains controversial.
Recommendations
2.2. For unilateral RVT in the absence of renal
impairment or extension into the IVC, we suggest either (1) supportive care with radiologic
monitoring for extension of thrombosis (if extension occurs, we suggest anticoagulation) or
(2) anticoagulation with UFH/LMWH or LMWH
in therapeutic doses rather than no therapy. If
anticoagulation is used, we suggest a total duration of between 6 weeks and 3 months rather
than shorter or longer durations of therapy
(Grade 2C). For unilateral RVT that extends
into the IVC, we suggest anticoagulation with
UFH/LMWH or LMWH for a total duration of
between 6 weeks and 3 months (Grade 2C).
2.3. For bilateral RVT with evidence of renal
impairment, we suggest anticoagulation with
UFH/LMWH or initial thrombolytic therapy with
tPA followed by anticoagulation with UFH/LMWH
(Grade 2C).
2.4-2.5 CVAD Prophylaxis in Neonates
In discussing anticoagulant CVAD prophylaxis, one
must consider two separate scenarios. The first is the
use of infusions or locks to keep the CVAD patent,
and the second is the use of systemic anticoagulation
to prevent large-vessel thrombosis.
A 2008 Cochrane review of peripherally placed
central venous catheters in neonates confirms the
value of UFH continuous infusion at 0.5 units/kg per h
compared with no treatment.284 This review considered two eligible randomized trials,285 which included
267 neonates. There was reduced risk of catheter
occlusion (typical risk ratio [RR], 0.28; 95% CI,
0.15-0.53; number needed to treat, 5; 95% CI, 3-8)
and no difference in the duration of catheter patency;
however, one study evaluated time to catheter
removal, censoring patients whose catheter was
removed because of therapy completion or death,
and identified benefit with heparin (adjusted hazard
ratio, 0.55; 95% CI, 0.36-0.83).285 This finding could
be due to a higher incidence of elective removal of
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e761S

catheters in neonates at the completion of therapy


in the heparin group (63% vs 42%; P 5 .002).285 The
other study detected no statistically significant differences between heparin and placebo when evaluating
time to blockage (P 5 .3) and time to sepsis (P 5 .1)
separately.286 No statistically significant differences
were observed in the risk of thrombosis (RR, 0.93;
95% CI, 0.58-1.51), catheter-related sepsis (RR, 1.96;
95% CI, 0.50-7.60), or new appearance or extension
of IVH (RR, 0.87; 95% CI, 0.25-3.03).284 A subsequent randomized controlled trial (RCT) demonstrated reduced infection with 0.5 units/mL heparin
through a neonatal long line compared with no heparin (RR, 0.57; 95% CI, 0.32-0.98; number needed to
treat, 9; 95% CI, 5-212).287
The local administration of thrombolytics is effective in restoring patency to blocked vascular catheters.
Both tPA and urokinase have been shown to restore
patency in 50% to 90% of CVADs; tPA has been
increasingly used relative to urokinase in children
during the past decade in doses ranging from 0.1 to
2 mg. Studies have included few neonates.239-248
There are no published studies comparing the use
of intermittent local thrombolysis as primary prophylaxis for CVAD patency. There are no studies that
consider the use of systemic heparin or LMWH as
primary prophylaxis for CVADs in neonates as distinct from older children.
Recommendation
2.4. For neonates with CVADs, we recommend
to maintain CVAD patency with UFH continuous
infusion at 0.5 units/kg per h over no prophylaxis (Grade 1A) or intermittent local thrombolysis (Grade 2C). For neonates with blocked
CVADs, we suggest local thrombolysis after
appropriate clinical assessment (Grade 2C).
2.6 Thromboprophylaxis for Blalock-Taussig
Shunts and Modified Blalock-Taussig Shunts
Blalock-Taussig shunts (subclavian-to-pulmonary
artery shunt) are a form of palliative surgery used
to enhance pulmonary artery blood flow. Modified
Blalock-Taussig shunts (MBTSs), in which a plastic
(Gortex; W. L. Gore & Associates, Inc) tube graft is
taken from the side of the subclavian artery and anastomosed to the pulmonary artery, are now standard
therapy.288 Thrombotic occlusion of MBTS has an
incidence of 1% to 17%. Smaller shunt size, smaller
infant size, and increased perioperative hemoglobin
level are risk factors for occlusion of MBTS within
24 h.289-291
Postdischarge occlusion is also reported.290,292 In a
study of 146 infants aged 60 days who underwent
MBTS and were discharged from the hospital alive,
e762S

21 (14%) died after discharge.292 Autopsies in 15 children attributed death to shunt thrombosis in five
infants (33%) and to myocardial infarction in two
(13%). The mortality of patients discharged on aspirin (11%) was almost identical to that of patients
discharged on no antithrombotic therapy (12.3%).292
Wells et al291 described the histologic appearance
of 155 MBTSs electively taken down at a mean of
8 months of age and reported that 21% had . 50%
stenosis; however, the role of thrombosis vs myofibroblastic proliferation in causing the obstruction was unclear.
A retrospective series of 546 MBTS procedures
reported no significant differences between heparin
and no heparin in early failure rate (1.4% vs 3.4%,
P 5 .29), in later failure rate (9.1% vs 13.6%, P 5 .17),
or between aspirin and no aspirin (11.0% vs 6.7%,
P 5 .18),293 and Li et al294 reported reduced thrombosis in a large cohort of patients treated with aspirin
for 12 months after shunt surgery. In another, much
smaller case study, aspirin was reported to decrease
the incidence of stent thrombosis after MBTS surgery.295 Mullen et al296 reported the safety of MBTS
without postoperative heparin therapy. No data support the use of ongoing anticoagulant therapy (heparin, LMWH, or VKAs) after MBTS surgery. No
published RCTs guide the antithrombotic medical
management of patients with MBTSs.
Recommendation
2.6. For neonates and children having MBTS, we
suggest intraoperative UFH therapy (Grade 2C).
For neonates and children after MBTS surgery,
we suggest either aspirin or no antithrombotic
therapy as compared with prolonged LMWH
or VKAs (Grade 2C).
2.7 Therapy for Blocked Blalock-Taussig
Shunts and MBTSs
Options for acute management of a thrombosed
MBTSs include reoperation with shunt takedown
and replacement, balloon angioplasty with or without
percutaneous catheter thrombectomy, or thrombolysis.297-299 There are insufficient data to recommend
any one specific therapy over another.
2.8 Thromboprophylaxis for Stage 1 Norwood
Procedures
Given that the standard Norwood procedure
involves an MBTS and that the shunt size often is
small (3.0 or 3.5 mm), prophylactic antithrombotic
recommendations following Norwood procedure are
based on those for MBTSs.300,301 Whether a different
strategy is required for Sano modifications of Norwood procedure is unknown.302
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

2.9-2.10 Therapy for Femoral Artery Thrombosis


in Neonates and Children

2.11 Prophylaxis for Peripheral Arterial


Catheters in Neonates and Children

Femoral artery thrombosis is most commonly seen


as a complication of cardiac catheterization (CC).
Few studies have considered neonates and children
separately. Short-term consequences of femoral artery
thrombosis include threatened limb viability and morbidity associated with anticoagulant or thrombolytic
therapy. Long-term consequences of femoral artery
thrombosis include leg-length discrepancies, muscle
wasting, claudication, and loss of arterial access.303-306
Symptomatic ischemia may occur at times when the
child experiences rapid growth, as occurs in the first
year of life and during puberty.305
Descriptions of treatment of femoral artery thrombosis in neonates or older children with thrombolytic
therapy, anticoagulation, thrombectomy or observation (no active therapy) consist exclusively of case
series without comparison groups.238,307-322 The general practice in the majority of childrens hospitals is
to initiate therapy with UFH for patients with postcardiac catheter femoral artery thrombosis. Previous
studies have reported that 70% of thromboses will
resolve with UFH alone.238 A recent report suggested
that LMWH may be a safe alternative in this situation, although the possible need for thrombolysis
or surgery makes LMWH a less attractive option.103
If , 4 h have passed since the CC-related UFH bolus
was given, then UFH may be commenced as a continuous infusion without a bolus. In acute limb-threatening
thrombosis, indirect evidence from adults supports
the use of thrombolytic or surgical interventions.

The majority of studies in this area have examined


interventions to prolong catheter patency as distinct from avoiding occlusive arterial thrombosis.323-326
Studies in pediatric intensive care have not separated
children from neonates. Catheter patency was significantly prolonged for UFH concentration of 5 units/mL
compared with 1 unit/mL,323 UFH with normal saline
vs dextrose flushes,324 and papaverine-supplemented
compared with placebo-supplemented solutions326
(Table 7).

Recommendations
2.9. For neonates and children with acute femoral
artery thrombosis, we recommend therapeutic
doses of IV UFH as initial therapy compared
with aspirin or no therapy (Grade 1B) or LMWH
(Grade 2C). We suggest subsequent conversion
to LMWH, or else continuation of UFH, to complete 5 to 7 days of therapeutic anticoagulation
as compared with a shorter or longer duration
(Grade 2C).
2.10. For neonates and children with limbthreatening or organ-threatening (via proximal
extension) femoral artery thrombosis who fail
to respond to initial UFH therapy and who have
no known contraindications, we recommend
thrombolysis (Grade 1C). For neonates and children with femoral artery thrombosis, we recommend surgical intervention compared with UFH
therapy alone when there is a contraindication to
thrombolytic therapy and organ or limb death is
imminent (Grade 1C).
www.chestpubs.org

Recommendation
2.11. For neonates and children with peripheral
arterial catheters in situ, we recommend UFH
continuous infusion at 0.5 units/mL at 1 mL/h
compared with normal saline (Grade 1A).
2.12 Therapy for Peripheral Artery
Thrombosis Secondary to Peripheral Artery
Catheters in Neonates and Children
Peripheral artery thrombosis in neonates and children is almost always due to arterial puncture or catheters.327 Tarry et al328 identified 44 cases of peripheral
arterial TEs secondary to catheterizations or arterial
punctures in children with nephrotic syndrome.
Friedman et al309 reported the use of microvascular
surgery in a heterogeneous group of neonates with
vascular injury. Albisetti et al329 reported 54 arterial
thromboses in 51 children of whom 96% had peripheral artery thrombosis. One case series of predominantly neonates proposed an algorithm to assist in
determining the role of surgery vs thrombolysis and
anticoagulation.263
Recommendation
2.12. For neonates and children with a peripheral arterial catheter-related TE, we suggest
immediate removal of the catheter (Grade 2B).
For neonates and children with a symptomatic
peripheral arterial catheter-related TE, we
suggest UFH anticoagulation with or without
thrombolysis or surgical thrombectomy and microvascular repair with subsequent heparin therapy (Grade 2C).
2.13-2.14 Prophylaxis of Umbilical Arterial
Catheters in Neonates
The incidence of symptomatic thrombosis of umbilical arterial catheters (UACs) is 1% to 3%,330-332 with
studies using sequential imaging and autopsy data
reporting a higher incidence.333,334 Factors increasing
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e763S

Catheter removal because


of malfunction:
1. 76%
2. 52%
P , .01
Patients aged , 1 y;
radial arterial
catheters
Retrospective
cohort
Selldn et al325/1987

NS 5 not significant. See Table 1 legend for expansion of other abbreviation.

1. n 5 296 catheters
2. n 5 42 catheters

Geometric mean of hours:


1. 33.5 h
2. 40.8 h
3. 43.5 h
Mean days:
1. 2.8 d
2. 6.3 d
P , .001
1. n 5 164
2. n 5 152
3. n 5 154

Heparin concentration/infusion rate:


1. 1 unit/mL, 1 mL/h
2. 1 unit/mL, 2 mL/h
3. 5 unit/mL, 1 mL/h
1. Intermittent flushing
2. Continuous infusion
RCT
Butt et al323/1987

Children

RCT
Rais-Bahrami et al324/1990

Newborns

Infusion 1-2 mL/h heparin 1 unit/mL:


1. heparinized normal saline
2. heparinized 5% dextrose water

1. n 5 30
2. n 5 30

Mean hours:
1. 78.1 h
2. 44.8 h
NS

Catheter failure:
1. 22%
2. 7%
P 5 .02
Mean hours for infants requiring
premature removal of catheters:
1. n 5 16, 107 h
2. n 5 21, 39 h
P , .001
Nonelective removal:
2 vs 1, NS
3 vs 1, P, .002
Time until catheter failure was
longer in patients receiving
papaverine (log-rank, P 5 .02)
1. n 5 124
2. n 5 115
1 unit/mL heparin:
1. placebo
2. papaverine (60 mg/500 mL)
Ages 3 wk to 18 y
RCT
Heulitt et al /1993

Comments
Duration of Catheter Patency
No. Patients
Groups
Age
Design
Study

326

Table 7[Recommendation 2.11] Prophylaxis for Peripheral Arterial Catheters in Neonates and Children

e764S

risk include longer catheter duration333 and the positioning of the UAC tip, which are routinely described
as high or low. The high position is at the level of the
T6 to T9 thoracic vertebral bodies. In this position,
the catheter tip is placed above the celiac axis,
superior mesenteric artery, and renal arteries and is,
therefore, above the diaphragm. The low position is
at the level of the L3 to L4 lumbar vertebral bodies,
and the position is therefore below these major vessels but above the aortic bifurcation. Barrington285
conducted a systematic review of five RCTs and
one alternate-assignment trial comparing outcomes
of clinical ischemic events, aortic thrombosis, IVH,
mortality, NEC, hypertension, and hematuria. Clinical ischemic events (RR, 0.53; 95% CI, 0.44-0.63;
consistent across five studies included) and aortic
thrombosis (RR, 0.31; 95% CI, 0.11-0.86; one study
with 62 infants) were less likely with high vs low
UAC placement; other outcomes were not significantly different between high and low placement.285
The short-term consequences of UAC-related thrombosis depend on the extent of the thrombosis but
include lower-limb ischemia,330,332,335 congestive cardiac failure, impaired renal function, and hypertension.336 Embolic events are also reported, and UAC
thrombosis has been linked to the development of
NEC.337,338 Longer-term outcomes include death,339
persistent renovascular hypertension, and lower-limb
growth abnormalities.340
Six RCTs addressed the use of low-dose heparin
infusions in neonates with UACs,341-346 which was the
subject of a systematic review by Barrington.347 Five
studies compared the use of UFH in the UAC infusate
with or without additional UFH in flush solutions
vs no UFH, whereas one compared the use of UFH
in the infusate vs UFH in the flush solution. End
points assessed in these studies included catheter
patency, aortic occlusion, other ischemic events, coagulation abnormalities, IVH, and hypertension. Two
studies used objective imaging to assess the incidence
of thrombosis.344,345 A reduced incidence of catheter
occlusion was consistent in the studies (four used UFH
1 unit/mL, and one used UFH 0.25 units/mL341), with
a pooled relative risk of 0.19 (95% CI, 0.10-0.33).
Despite the consistent reduction in catheter occlusion, none of these studies were able to demonstrate
that UFH had any effect on aortic thrombosis or
other ischemic events. There was also no significant
difference in the incidence of IVH.
McDonald et al348 reported the use of full systemic
UFH vs low-dose UFH in an RCT that included only
19 infants. Although there was a trend toward a
reduced incidence of aortic thrombosis, this did not
achieve statistical significance, and data on the incidence of IVH and other coagulation abnormalities
were not reported.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

The association between the use of UFH and the


occurrence of IVH in preterm neonates remains
controversial. In the review by Barrington,347 no
association between heparin exposure and IVH was
identified. However, five of six studies in this review
included infants of various gestational ages, some of
whom may have been at relatively low risk for IVH,
contrasting with other published data.349,350 Lesko et al349
reported a fourfold increase in IVH in low-birthweight infants in a case control study; however, the
CIs were relatively wide (OR, 3.9; 95% CI, 1.4-11.0).
In addition, the median birth weight was lower in
the UFH group, which also had a higher incidence
of concomitant illnesses. Malloy and Cutter,350 also
reported higher UFH exposure in low-birth-weight
infants with IVH. Again, confounding factors related
to the severity of illness were not included in the
model used for analysis.
Recommendations
2.13. For neonates with UACs, we suggest UAC
placement in a high rather than low position
(Grade 2B).
2.14. For neonates with UACs, we suggest prophylaxis with a low-dose UFH infusion via the
UAC (heparin concentration of 0.25-1 unit/mL,
total heparin dose of 25-200 units/kg per day) to
maintain patency (Grade 2A).
2.15 Treatment of Aortic Thrombosis
Non-UAC-associated or spontaneous aortic thrombosis is a rare event.351-361 Many published cases have
evidence of extensive thrombosis involving either the
abdominal or the thoracic aorta, the later sometimes
mimicking coarctation at presentation.362 The outcome of these events is variable, but overall mortality
appears to be relatively high.
A review summarized the management of aortic
thrombosis.363 Therapeutic options include heparin
or LMWH, thrombolytic therapy, and surgical throm
bectomy.310,335,336,352,358,361,362,364-371 There are insufficient
data to recommend any one treatment over others.

is 40%,373,374 with younger children (ie, aged , 10 years)


having an increased incidence compared with older
children.373,374 Patient size, patient hemodynamic status,
operator technique, larger catheter size, total time
of arterial cannulation, and procedural vs diagnostic
catheters are all factors that affect the risk for arterial
thrombosis.313
Taylor et al306 described 58 children aged , 5 years
at the time of catheterization and who were evaluated
5 to 14 years later using arterial duplex scanning
and lower-extremity radiographs of bone length.
Arterial occlusion was present in 33% of patients.
Celermajer et al303 reported that . 30% of previously
catheterized children and adolescents presented with
vascular access problems at subsequent catheterizations because of an occluded vessel, a stenosed vessel,
or scar tissue.
Five prospective trials examined the value of prophylaxis to prevent femoral artery thrombosis.373-377
Freed et al373 demonstrated that prophylactic anticoagulation therapy with aspirin does not significantly
reduce the incidence of femoral artery thrombosis.
Anticoagulation therapy with 100 to 150 units/kg UFH
reduces the incidence from 40% to 8%. A randomized trial of 366 children suggested that a 50-unit/kg
bolus of heparin may be as effective as a 100 units/kg
when given immediately after arterial puncture
(9.8% vs 9.3% incidence of arterial thromoboses);
however, the CIs around the estimate are wide
(0.6%; 95% CI, 25.5%-6.6%).377
Recommendation
2.16. For neonates and children requiring CC
via an artery, we recommend administration
of IV UFH as thromboprophylaxis over no prophylaxis (Grade 1A) or aspirin (Grade 1B). For
neonates and children requiring CC via an
artery, we recommend the use of UFH doses
of 100 units/kg as a bolus compared with a
50-unit/kg bolus (Grade 1B). In prolonged procedures, we suggest further doses of UFH rather
than no further therapy (Grade 2B).
2.17 Cerebral Sinovenous Thrombosis in Neonates

2.16 Prophylaxis for Cardiac Catheterization


in Neonates and Children
The femoral artery is the most common access
site for CC in neonates and children, although
radial access has been reported in older children.372
Although a number of studies have analyzed infants
(aged , 1 year) compared with older children, the
available data do not allow separate consideration of
neonates from children.
The incidence of femoral artery thrombosis in
the absence of any thromboprophylaxis after CC
www.chestpubs.org

The incidence of cerebral sinovenous thrombosis


(CSVT) in neonates is at least 2.6 per 100,000.378
Premature and term neonates are affected,379 and
CSVT can occur antenatally.380 Seizures and lethargy
are frequent, and focal neurologic deficits rare.381,382
Venous infarcts are present in . 50%, of which the
majority are hemorrhagic.383,384 IVH is also frequent. Among term neonates with IVH, underlying
CSVT is documented in nearly one-third (31%) and
is more likely when thalamic hemorrhage is present
(P 5 .03).385
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e765S

Reported outcomes after neonatal CSVT include


death in 7% to 19% and neurologic impairments in
36% to 79% of survivors.383,384,386 Adverse neurologic
outcomes include cognitive and motor deficits and in
20% to 40%, epilepsy.387 The presence of infarcts at
diagnosis and perinatal complications predict worse
outcome.388,389
Although overt, recurrent CSVT is rare in neonates, propagation of the initial thrombus after diagnosis of CSVT is a concern. A cohort study reported
asymptomatic propagation in the first week after
diagnosis in 11 of 44 (25%) neonates treated without
anticoagulation.390
There are no RCTs391; however, data on the
safety of anticoagulation in neonates with CSVT are
available.378,383,384,392 There are significant geographic
differences in physician decisions to treat neonates
with CSVT with anticoagulants, with European and
Canadian physicians much more likely to treat than
US physicians.392 In a consecutive cohort treatment
safety study using standardized protocols for anticoagulation of neonatal CSVT, bleeding occurred in
three of 37 (8%) treated neonates but was not fatal
in any.390 A subsequent report from the same authors
reported nonfatal ICH in 14% of neonates with pretreatment ICH and only 2% in those without pretreatment ICH.393 The most common anticoagulant
used is LMWH.386,392,394 LMWH has been reported to
be safe even in the presence of significant thalamic
hemorrhage.395
The optimal dose and duration of anticoagulant treatment is not known. However, neonates recanalize
faster than older children, and the rate of recanalization is greatest in the first 3 months after diagnosis.
About 50% of neonates have fully recanalized by
6 weeks to 3 months after diagnosis, and recanalization is observed in 65% by 6 months and 75% by
1 year.390 Therefore, one approach is to assess for
recanalization at 6 weeks and if complete, to stop
anticoagulants, or if incomplete, to continue for an
additional 6 weeks (3 months anticoagulation) and
then stop. Early neurosurgical intervention may be
necessary for even mild ventriculomegaly due to
obstructive hydrocephalus.

2.18-2.20 Arterial Ischemic Stroke in Neonates


The diagnosis of neonatal arterial ischemic stroke
(AIS) is very challenging because the clinical presentation is nonspecific, and routine imaging studies may
miss AIS in neonates.396 MRI with diffusion-weighted
imaging usually is diagnostic for AIS.397,398
The incidence of acute neonatal AIS is one in
4,000 live births.399 A form of perinatal stroke with
delayed diagnosis has been referred to as presumed
perinatal AIS.400,401 Such infants are neurologically
normal in the first month of life, demonstrate early
hand preference or other signs of hemiparesis typically between 4 and 12 months of age, and have a
CT scan showing a remote lesion consistent with prenatal or perinatal AIS. A variety of risk factors have
been described for term and preterm neonates.402-406
In term infants with AIS, the typical distribution is
the middle cerebral artery, more commonly left than
right, and small artery infarcts can occur. Hemorrhagic
conversion is well recognized.
Neurologic deficits or epilepsy occur in 50% to
75% of survivors; sensorimotor deficits and feeding
problems are most common.400,407,408 Outcomes may be
worse in preterm neonates.409 Long-term follow-up is
critical because later deficits with brain maturation
often emerge.410 Radiographic features may predict
outcomes.411,412 Recurrent stroke is very rare after AIS
in the neonatal period.413-415
Recommendations
2.18. For neonates with a first AIS in the absence
of a documented ongoing cardioembolic source,
we suggest supportive care over anticoagulation or aspirin therapy (Grade 2C).
2.19. For neonates with a first AIS and a documented cardioembolic source, we suggest anticoagulation with UFH or LMWH (Grade 2C).
2.20. For neonates with recurrent AIS, we suggest anticoagulant or aspirin therapy (Grade 2C).

Recommendation
2.17. For neonates with CSVT without significant ICH, we suggest anticoagulation, initially with UFH or LMWH and subsequently
with LMWH, for a total therapy duration between
6 weeks and 3 months rather than shorter or
longer treatment duration (Grade 2C). For neonates with CSVT with significant hemorrhage,
we suggest either (1) anticoagulation or (2) supportive care with radiologic monitoring of the
e766S

thrombosis at 5 to 7 days and anticoagulation if


thrombus extension is noted as compared with
no therapy (Grade 2C).

2.21 Neonates With Purpura Fulminans


Purpura fulminans is an acute, lethal syndrome of
disseminated intravascular coagulation characterized
by rapidly progressive hemorrhagic necrosis of the
skin due to dermal vascular thrombosis.416-419 The skin
lesions start as small, ecchymotic areas that increase
in a radial fashion, become purplish black with bullae,
and then turn necrotic and gangrenous. The lesions
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

occur mainly on the extremities but can occur on the


buttocks, abdomen, scrotum, and scalp.
The syndrome is due to homozygote protein C (most
commonly) or protein S deficiency or compound heterozygous states with undetectable plasma levels of
the respective protein.420-434 Rarely have other causes
been described.435 The classic clinical presentation
consists of cerebral or ophthalmic damage (or both)
that occurred in utero. In up to 70% of cases, purpura
fulminans occurs within hours or days of birth, and,
on rare occasions, large-vessel thrombosis occurs.
The diagnosis is based on the appropriate clinical picture; a very-low or undetectable protein C/protein S
level; heterozygous deficiency of the same protein in
the parents; and, ideally, identification of the molecular defect. The presence of very-low levels of protein
C/protein S in the absence of clinical manifestations
and of a family history cannot be considered diagnostic because physiologic plasma levels can be as low as
0.12 units/mL.
Although clinicians have used numerous forms of
initial therapy, 10 to 20 mL/kg of FFP every 6 to 12 h
is usually the form of therapy that is most readily
available.436,437 Plasma levels of protein C achieved
with these doses of FFP vary from 15% to 32% at
30 min after the infusion and from 4% to 10% at
12 h.426 Plasma levels of protein S (which is entirely
bound to C4b) are 23% at 2 h and 14% at 24 h, with
an approximate half-life of 36 h.438,439 Doses of protein
C concentrate have ranged from 20 to 60 units/kg.
In one study, a dose of 60 units/kg resulted in peak
protein C levels of . 0.60 units/mL.426 Replacement
therapy should be continued until all the clinical
lesions resolve, which is usually at 6 to 8 weeks. In
addition to the clinical course, plasma D-dimer concentrations may be useful for monitoring the effectiveness of protein C replacement.
The modalities used for the long-term management of infants with homozygous protein C/protein S
deficiency include oral anticoagulation therapy, replacement therapy with either FFP or protein C concentrate, and liver transplantation.419,437,440 To avoid skin
necrosis when oral anticoagulation therapy is initiated,
replacement therapy should be continued until the
INR is therapeutic. The optimal therapeutic range is
unknown; ranges from 2.5 to 4.5 have been reported.
The risks of oral anticoagulation therapy include bleeding with high INRs and recurrent purpuric lesions
with low INRs. Frequent monitoring of INR values
is required if these complications are to be avoided.
Recommendation
2.21. For neonates with clinical presentations
of homozygous protein C deficiency, we recommend administration of either 10 to 20 mL/kg
www.chestpubs.org

of FFP every 12 h or protein C concentrate,


when available, at 20 to 60 units/kg until the
clinical lesions resolve (Grade 1A). For neonates with homozygous protein C deficiency,
after initial stabilization, we recommend longterm treatment with VKA (Grade 1C), LMWH
(Grade 1C), protein C replacement (Grade 1B),
or liver transplantation (Grade 1C) compared
with no therapy.
2.22 DVT and PE in Children
Unlike adults, 95% of VTEs in children are secondary to an identifiable risk factor.13,22,151,441-443 The
most common risk factor is the presence of a
CVAD.13,22,441,444-447 When spontaneous thrombosis
occurs in children, it is usually in the lower limbs.17
Recurrent VTEs occur in 7.5% of children.13,17,22,441
Prospective studies reveal that asymptomatic central venous line-related thrombosis occur frequently in
children.113,445,448 Radiographically confirmed asymptomatic CVAD-related thromboses in children are
of clinical importance for a number of reasons.
First, CVAD-related VTEs are associated with CVADrelated sepsis.447 Second, CVAD-related thrombosis
is a common source for PE in children,17,449,450 which
may be fatal.446 Third, recurrent CVAD-related clot
may result in loss of venous access that may be
necessary for life-saving intervention such as organ
transplantation.17,22,451 Finally, many children have persistent right-to-left intracardiac shunts through which
paradoxical embolism may occur.17,446
PTS occurs in 12% to 65% of children following
venous thrombosis.451,452 Although treatment studies
have included this as an outcome measure, delay
in initial treatment and recurrent thrombosis are
reported risk factors.452
There has been one multicenter randomized
trial of anticoagulation for VTE in children.151 The
REVIVE (Reviparin in Venous Thromboembolism)
trial randomized children (aged . 3 months) with a
first VTE to receive either UFH and then VKAs (target INR, 2.5) for 3 months or an LMWH (reviparin)
adjusted to achieve a target anti-Xa level of 0.5 to
1.0 units/mL for 3 months. The study was closed early
because of slow recruitment prior to completion of
target recruitment. As a result, the study failed to
demonstrate or exclude a reduction or increase in
recurrence (OR, 0.53; 95% CI, 0.05-4.00) or bleeding
(OR, 0.41; 95% CI, 0.04-2.76).
Table S1 summarizes the other studies evaluating treatment of venous thrombosis in children
(see Acknowledgments for more information on
accessing this supplemental table). Many of the recommendations rely on indirect evidence for treatment
of DVT and PE in adults.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e767S

Recommendations
2.22.1. In children with first VTE (CVAD and
non-CVAD related), we recommend acute anticoagulant therapy with either UFH or LMWH
(Grade 1B). We recommend initial treatment
with UFH or LMWH for at least 5 days (Grade 1B).
For ongoing therapy, we suggest LMWH or
UFH. For patients in whom clinicians will subsequently prescribe VKAs, we recommend
beginning oral therapy as early as day 1 and discontinuing UFH/LMWH on day 6 or later than
day 6 if the INR has not exceeded 2.0 compared
with no therapy (Grade 1B).
2.22.2. We suggest that children with idiopathic
VTE receive anticoagulant therapy for 6 to
12 months compared with no therapy (Grade 2C).
Underlying values and preferences: Families who
place a high value on avoiding the unknown risk of
recurrence in the absence of an ongoing risk factor
and a lower value on avoiding the inconvenience of
therapy or potential impact of therapy on growth and
development and bleeding risk associated with antithrombotic therapy are likely to choose to continue
anticoagulant therapy beyond 6 to 12 months.
2.22.3. In children with secondary VTE (ie, VTE
that has occurred in association with a clinical
risk factor) in whom the risk factor has resolved,
we suggest anticoagulant therapy be administered for 3 months (Grade 2C) as compared
with no further therapy. In children who have
ongoing, but potentially reversible risk factors,
such as active nephrotic syndrome or ongoing
asparaginase therapy, we suggest continuing anticoagulant therapy beyond 3 months in either
therapeutic or prophylactic doses until the risk
factor has resolved (Grade 2C).
2.22.4. In children with recurrent idiopathic
VTE, we recommend indefinite treatment with
VKAs (Grade 1A).
2.22.5. In children with recurrent secondary
VTEs with an existing reversible risk factor for
thrombosis, we suggest anticoagulation until
resolution of the precipitating factor but for a
minimum of 3 months as compared with no further therapy (Grade 2C).
2.22.6. In children with a CVAD in place who
have a VTE, if a CVAD is no longer required, or
is nonfunctioning, we recommend it be removed
(Grade 1B). We suggest at least 3 to 5 days of
anticoagulation therapy prior to its removal
e768S

rather than no anticoagulation prior to removal


(Grade 2C). If CVAD access is required, and
the CVAD is still functioning, we suggest that
the CVAD remain in situ and the patient be
given anticoagulants (Grade 2C). For children
with a first CVAD-related VTE, we suggest initial management as for secondary VTE as previously described.
2.22.7. In children with CVAD in place who have
a VTE and in whom the CVAD remains necessary,
we suggest after the initial 3 months of therapy,
that prophylactic doses of VKAs (INR range,
1.5-1.9) or LMWH (anti-Xa level range, 0.10.3 units/mL) be given until the CVAD is removed
(Grade 2C). If recurrent thrombosis occurs while
the patient is receiving prophylactic therapy,
we suggest continuing therapeutic doses until
the CVAD is removed and for a minimum of
3 months following the VTE (Grade 2C).
2.23 Thrombolysis in Pediatric Patients With DVT
A number of case series have described thrombolysis for DVT in children.222,226,235-238,329,453-460 A retrospective cohort found improved outcomes with
respect to PTS when children received thrombolysis
rather than anticoagulation alone.461 Thrombolysis
has been given systemically as catheter-directed
doses, alone, and in combination with mechanical
thrombolysis.250,461-464 A number of reviews have summarized the experience of catheter-directed and
systemic thrombolysis for DVT in children.219,465,466
The risk-benefit ratio in terms of improved thrombosis
outcome vs bleeding risk remains uncertain, as does
the optimal dose and delivery technique. On the basis
of adult data, the use of thrombolysis may be best
restricted to limb- or life-threatening thrombosis.467
Recommendation
2.23. In children with VTE, we suggest that
thrombolysis therapy be used only for life- or
limb-threatening thrombosis (Grade 2C). If thrombolysis is used in the presence of physiologically
low levels or pathologic deficiencies of plasminogen, we suggest supplementation with plasminogen (Grade 2C). In children with VTE in
whom thrombolysis is used, we suggest systemic
thrombolysis or catheter-directed thrombolysis
depending on institutional experience and, in
the latter case, technical feasibility.
2.24 Thrombectomy and IVC Filter Use
in Pediatric Patients With DVT
Case reports and small case series in children
report the use of thrombectomy for massive VTE or
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

PE468-471 or for life-threatening thrombosis (Fontan


circuit).270,311,314,319,469,471,472 Recent reviews have also
described both open and percutaneous methods of
thrombectomy.465,466 Thrombectomy may be an appropriate first-line therapy in children postcardiac surgery who have significant thrombus in the surgical
field, especially if a shunt is acutely compromised.
Reports of successful and failed IVC filters in
children have been published.253,254,256-258,260,469,473-476
Raffini et al reported,259 in a prospective cohort study,
that 5% of children with DVT required IVC filters.
The filter usually is placed through the femoral or
jugular approach and may remain in situ for life or
may be temporary. Removable filters are preferable,
and they may remain in situ for up to 5 months.258
Vena cava filter placement is restricted to children
who weigh . 10 kg because of the size of the IVC and
the available filters. In addition, the availability of
a skilled pediatric interventional radiologist with
experience in this field will be a major determinant
of the risk-benefit ratio in individual patients. The
complications of filter placement include extension
of preexisting thrombosis up to the level of the filter,
thrombus formation within the filter basket, and perforation of the IVC.477-479
Recommendation
2.24. In children with life-threatening VTE,
we suggest thrombectomy (Grade 2C). In children who have had a thrombectomy, we suggest
anticoagulant therapy as per recommendation (Recommendations 2.22) (Grade 2C). In children . 10 kg body weight with lower-extremity
VTE and a contraindication to anticoagulation,
we suggest placement of a retrievable IVC filter
(Grade 2C). In children who receive a filter, we
suggest that the filter be removed as soon as
possible if thrombosis is not present in the basket of the filter and when contraindication to
anticoagulation is resolved (Grade 2C). In children who receive an IVC filter, we recommend
appropriate anticoagulation for VTE (see Recommendation 1.2) as soon as the contraindication to anticoagulation is resolved (Grade 1C).
2.25 DVT in Children With Cancer
One RCT (the REVIVE study) compared LMWH
and UFH/warfarin in the treatment of DVT in 78 children, but only 30% were cancer patients.151 The
CIs were consistent with substantial benefit and
substantial detriment. LMWH is the preferred anticoagulant in many pediatric cancer patients because
of the ease of maintaining the anticoagulation therapy
around the usual frequent procedures, such as lumbar
punctures.480
www.chestpubs.org

The rates of thrombosis in childhood cancer are much


lower than in adults.480-487 Furthermore, many childhood cancers have high cure rates, so active cancer may
not be an ongoing factor once treatment is under way.
Finally, therapy for childhood cancer is often intense
and associated with significant thrombocytopenia,
increasing the bleeding risks of anticoagulant therapy.488
Thus, children with cancer and VTE may not benefit
from antithrombotic therapy beyond 3 months, providing that other risk factors have resolved.489
There is only one case report of a pediatric patient
with cancer developing thrombosis before the diagnosis of cancer.490 This suggests that other risks factors
play a significant role in the development of VTE in
this population. For example, the risk of VTE in children with cancer varies considerably with the chemotherapeutic protocol being used.487,491 The presence
of CVADs also appears to greatly increase the rate of
thrombosis.486,487,491 Further, specific host factors may
be important.492 Thus, although the optimal duration
of therapy is unknown, the control of reversible risk
factors seems to be associated with a considerable
reduction in risk of thrombosis recurrence and, therefore, important to consider in determining the duration of therapy. However, there remains little direct
evidence to support recommendations.487,493
Recommendation
2.25. In children with cancer, we suggest that
management of VTE follow the general recommendations for management of VTE in children.
We suggest the use of LMWH in the treatment
of VTE for a minimum of 3 months until the precipitating factor has resolved (eg, use of asparaginase) (Grade 2C).
Remarks: The presence of cancer, the need for surgery, chemotherapy, or other treatments may modify
the risk-benefit ratio for treatment of VTE, and clinicians should consider these factors on an individual
basis.
2.26 Children With Antiphospholipid
Antibodies and DVT
Antiphospholipid antibodies (APLAs) are associated
with an increased risk of thrombosis in children,494-496
although whether this risk is similar to that of adults
remains uncertain.497-499 There is no direct evidence to
guide the optimal therapy for DVT in children with
APLAs or to support or refute the role of primary
prophylaxis.
Recommendation
2.26. For children with VTE in the setting
of APLAs, we suggest management as per
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e769S

general recommendations for VTE management


in children.
2.27 Children With DVT and Positive
Inherited Thrombophilia Testing
The impact of a variety of inherited thrombophilia
markers on the risk of recurrence for pediatric venous
thrombosis has long been debated and was the subject
of a meta-analysis500 and review article.501 The literature on which these reviews were based were cohort
and case-control studies as well as case series, and there
remain many questions about the optimal testing strategy and the impact of results on therapeutic decisions.
There are insufficient data to support the presence or
absence of thrombophilia markers as a determinant of
intensity or duration of therapy and as distinct from the
presence or absence of clinical precipitants of thrombosis (ie, spontaneous vs secondary thrombosis).
Recommendation
2.27. For children with VTE independent of the
presence or absence of inherited thrombophilic
risk factors, we suggest that the duration and
intensity of anticoagulant therapy as per Recommendation 2.22.
2.28 Children With VTE and Structurally
Abnormally Venous Systems
Structural venous abnormalities as precipitants of
venous thrombosis in childhood are well described,
although patients with such abnormalities often do not
present until adult life. In the lower venous system,
the most common abnormalities are interrupted or
duplex IVC, presumably secondary to disrupted embryonic development of the IVC itself.502-509 However, in
the upper venous system, thoracic outlet syndrome,
eponymously named Paget-Schroetter syndrome, are
believed to be due to chronic trauma to the subclavian
vein secondary to reduced anatomic space for the
vein usually as a result of an abnormal relationship
with the first rib, abnormal fibrous bands or muscle
development in athletes.510-519 Management of the
venous thrombosis and the underlying structural
abnormality has included acute anticoagulation; both
local and systemic thrombolysis through a number
of techniques; percutaneous angioplasty; thrombectomy; venous reconstruction; and in the case of PagetSchroetter syndrome, decompression of the thoracic
inlet through removal of relevant bone and muscle.
No randomized trials have been conducted.
Recommendation
2.28. For children with first VTE secondary
to structural venous abnormalities, we suggest
e770S

anticoagulation as per other spontaneous VTE


(Recommendations 2.22) and consideration of
subsequent percutaneous or surgical interventions depending on patient factors and institutional experience. For children with recurrent
VTE secondary to structural venous abnormalities, we suggest indefinite anticoagulation unless
successful percutaneous or surgical interventions
can be performed (Grade 2C).
2.29 Children With Right Atrial Thrombosis
Right atrial and intracardiac thromboses are most
commonly diagnosed in children who have CVADs
extending into the right atrium.273,520 The epidemiology and risk associated with right atrial thrombosis
appears to be different in children compared with
adults.273 The natural history appears to be resolution
irrespective of therapy, and many children are asymptomatic. Risk stratification based on clot size and
mobility appears to be useful.273,520 For low-risk patients
with a clot , 2 cm in size, nonmobile, and attached to
the atrial wall, not pedunculated or snake shaped,
then removal of the CVAD with or without anticoagulation appears appropriate. For high-risk thrombosis cases, systemic anticoagulation should be offered.
Thrombolysis or percutaneous or surgical thrombectomy have considerable risks and should be considered on an individualized basis.273
Recommendation
2.29. For children with right atrial thrombosis
related to CVAD, we suggest removal of the
CVAD with or without anticoagulation depending on the individual risk factors compared with
leaving the CVAD in situ (Grade 2C). For children
with large (. 2 cm), mobile, right atrial thrombosis, we suggest anticoagulation with appropriately
timed CVAD removal and consideration of surgical intervention or thrombolysis based on individualized risk-benefit assessment compared
with no anticoagulation therapy (Grade 2C).
2.30-2.34 Children With CVADs
Loss of CVAD patency most often is due to intraluminal occlusion with either thrombus or chemical
deposition, although it may be secondary to largevessel thrombosis involving the vein in which the
CVAD is situated. CVAD patency is necessary for
therapy to be effectively given through the CVAD.
Blocked CVADs may be at increased risk of infection
and lead to increased anesthetic and surgical exposure
when they require replacement. Primary prophylaxis
to maintain patency has usually considered intermittent CVAD heparin or saline flushes, continuous
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

CVAD heparin, or saline infusions. Taurolidine-citrate


locks have been compared with heparin locks and
shown to reduce rates of bloodstream infection.521
Restoration of CVAD patency has usually involved
intermittent bolus dosing of a variety of agents, with
failure to restore patency being defined as the CVAD
removal and replacement rate. Primary prophylaxis
to avoid large-vessel thrombosis around the CVAD
usually involves systemic anticoagulation and needs
to be considered separately.
Studies that have addressed the issue of CVAD
patency include a trial that evaluated the use of saline
vs combination saline and 1 unit/mL UFH in a singlecenter, blinded randomized clinical trial.522 The study
failed to demonstrate or exclude a beneficial or detrimental effect on CVAD patency between the two
groups (RR, 7.63; 95% CI, 0.4-144.9).
An unblinded, randomized crossover study in which
14 children received UFH 50 units/kg flush vs standard care every 12 h failed to demonstrate or exclude
a beneficial or detrimental effect on CVAD patency.523 A literature review by Kannan524 provided no
evidence to support heparin over normal saline as an
intermittent flush.
In contrast, a meta-analysis of studies in adults
with CVADs reported benefit of heparin compared
with saline in terms of thrombosis, bacterial colonization, and possible bacteremia.447 More than 40% of
children with cancer had CVAD occlusions despite
weekly heparin locks.525 In the hemodialysis setting,
alteplase 1 mg/mL was shown to be more effective
than heparin 5,000 units/mL in reducing intraluminal
clot between hemodialysis sessions.526 A multicenter
study of 577 pediatric cancer patients with CVADs
reported that urokinase administration every 2 weeks
reduced both CVAD occlusion rates and catheterrelated infections compared with heparin administration.527 Most other studies that have reported
the use of local thrombolytic agents reported therapy for CVAD blockage and, hence, restoration of
patency.241,243,246,528
Recommendation
2.30. For CVADs, we suggest flushing with normal saline or heparin or intermittent recombinant urokinase (rUK) to maintain patency
as compared with no therapy (Grade 2C). For
blocked CVADs, we suggest tPA or rUK to restore
patency (Grade 2C). If after at least 30 min
following local thrombolytic instillation CVAD
patency is not restored, we suggest a second
dose be administered. If the CVAD remains
blocked following two doses of local thrombolytic agent, we suggest radiologic imaging to
rule out a CVAD-related thrombosis (Grade 2C).
www.chestpubs.org

There are two RCTs reporting thromboprophylaxis of CVADs to prevent CVAD-related DVT.113
The PROTEKT Study randomized 186 children
aged 3 months with varying underlying conditions
to reviparin (n 5 92) (anti-Xa levels, 0.1-0.3 units/mL)
vs standard care (n 5 94) (up to 3 units/kg per h
UFH).113 The incidence of asymptomatic CVADrelated thrombosis was 14.1% (11/78) in the reviparin
group vs 12.5% (10/80) in the standard care group
(OR, 1.15; 95% CI, 0.42-3.23).113 The study was
closed early because of slow patient recruitment.
Schroeder et al47 reported a single-center RCT comparing 10 units/kg per h continuous UFH infusion
to placebo in 90 children aged , 1 year with CVADs
postcardiac surgery. There was no difference in CVADrelated thrombosis in the treatment or placebo groups
(15% vs 16%; difference of 1%; 95% CI, 214%-16%).
The study was closed after an interim analysis showed
futility.
The incidence of CVAD-related thrombosis varies
with the underlying patient population, and this has
led to some more-specific disease-related studies
to examine the role of primary prophylaxis. CVADrelated thrombosis is reported to be common in
children with cancer.484,485 There are two RCTs that
studied thromboprophylaxis in children with cancer.
The Prophylactic Antithrombin Replacement in Kids
with Acute Lymphoblastic Leukemia Treated with
Asparaginase (PARKAA) trial studied the use of antithrombin concentrate vs no therapy in 85 patients
with pediatric acute lymphoblastic leukemia (ALL)
treated with asparaginase.486 Seven of 25 (28%) patients
treated with antithrombin compared with 22 of 60
(37%) not treated had thrombosis (difference, 29%;
95% CI, 230%-13%), but the study was not powered
to show efficacy. Ruud et al529 studied the use of warfarin compared with no therapy in the prevention of
CVAD-related thrombosis in children with cancer.
The study enrolled 73 children, and 15 of 31 (48%)
on warfarin and 17 of 42 (40%) on no therapy developed a VTE (difference, 12%; 95% CI, 215%-31%).
The study was terminated without full recruitment
after an interim study showed futility.
Cohort studies and case series have also addressed
this issue. Elhasid et al107 found LMWH (mean dose,
0.84 mg/kg once daily) to be apparently safe when
compared with historical controls in preventing thrombosis in 41 patients with ALL. Nowak-Gttl et al530
gave LMWH (dose, 1 mg/kg once daily) as primary
thromboprophylaxis to children and adolescents with
Ewing sarcoma (n 5 36) and osteogenic sarcoma
(n 5 39). None of the patients developed TE complications during the postoperative period. Meister et al531
reported that enoxaparin (1 mg/kg per day) in addition to antithrombin supplementation reduced the
rate of thrombosis in children with ALL and CVADs
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e771S

being treated in the BFM (Berlin-Frankfurt-Mnster)


95 of 2,000 studies compared with antithrombin alone
(nine of 71 [13%] with antithrombin alone and zero
of 41 [0%] with enoxaparin alone; difference, 13%;
95% CI, 5%-20%). None of these series is adequate
to address the question of efficacy because of sample
size and study design.487

Recommendation
2.35. For children who have a BCPS, we suggest
postoperative UFH (Grade 2C).
2.36 Fontan Surgery

Recommendation
2.31. For children with short- or medium-term
CVADs, we recommend against the use of routine systemic thromboprophylaxis (Grade 1B).
The incidence of CVAD-related VTE in children
receiving long-term total parenteral nutrition (TPN)
varies from 1% based on clinical diagnosis to 35%
based on ventilation perfusion scans or echocardiography to 75% based on venography.444,532-538
Two studies have reported the use of VKA primary
prophylaxis in this group of patients.444,535 However,
VKA primary prophylaxis commonly is used for children receiving long-term home parenteral nutrition,
despite the lack of similar practice in adults.
Recommendation
2.34. For children receiving long-term home
TPN, we suggest thromboprophylaxis with VKAs
(Grade 2C).
2.35 Glenn Procedure or Bilateral
Cavopulmonary Shunt
Glenn successfully performed the classic cavopulmonary anastomosis in 1957 as palliation for tricuspid
atresia. The bidirectional Glenn procedure is now
frequently used as an intermediate step in patients
with single ventricles prior to definitive Fontan surgery (following Blalock-Taussig shunts in hypoplastic
right-side hearts and following stage I Norwood procedure in hypoplastic left-side hearts). Thrombotic
complications following the Glenn shunt procedure
are infrequent.539-544 No published data support
the need for routine thromboprophylaxis. However,
once again, the fact that many patients subsequently
proceed to Fontan procedures has led to some suggestions that thromboprophylaxis is warranted after a
Glenn shunt to reduce the risk of thrombosis in the
pulmonary vasculature, hence increasing the likelihood of successful conversion to a full Fontan circuit.
Current clinical practices vary, and include no anticoagulation, UFH followed by aspirin, and UFH followed by warfarin therapy. There is no evidence to
support a preference for any of these approaches.
Recommendations for patients undergoing a bilateral
cavopulmonary shunt (BCPS) procedure are theree772S

fore based on generalization from other major vascular procedures in infants and children.

The Fontan procedure, or a modified version, is


the definitive palliative surgical treatment of most
congenital univentricular heart lesions.545,546 TE remains
a major cause of early and late morbidity and mortality. Reported incidences of VTE and stroke ranged
from 3% to 16% and 3% to 19%, respectively, in retrospective cohort studies where thrombosis was the
primary outcome and from 1% to 7% in retrospective
studies assessing multiple outcomes.547,548 TE may
occur any time after Fontan procedures but often
present months to years later.549 No predisposing factors have been identified with certainty, although this
may be due to inadequate power and the retrospective nature of the studies.
Transesophageal echocardiography is more sensitive
than transthoracic echocardiography for the diagnosis
of intracardiac and central venous thrombosis.550-552
MRI has been reported to be useful in noncomparative
studies.553 Despite aggressive therapy, TE following
Fontan procedures carries a high mortality and responds
to therapy in , 50% of cases.554,555 There is no consensus
in the literature or in routine clinical practice about the
optimal type or duration of antithrombotic therapy to
prevent such events.271,542,556,557 Consequently, a wide
variety of prophylactic anticoagulant regimes are in use.
There are very few studies that compare treatment
options.547,548 Some cohort studies report anticoagulation or aspirin to be better than no therapy in terms
of thrombotic complications,558 although others question this finding.559 The only randomized trial performed compared aspirin (5 mg/kg per day) to initial
UFH followed by warfarin (target INR, 2.5; range,
2.0-3.0) as primary prophylaxis for 2 years postFontan surgery. The study found no difference in
thrombosis (cumulative thrombosis rates, 19% at
2 years) or bleeding.560 No studies comparing optimal
duration of therapy have been performed.
Recommendation
2.36. For children after Fontan surgery, we recommend aspirin or therapeutic UFH followed
by VKAs over no therapy (Grade 1C).
2.37 Endovascular Stents
Endovascular stents are used with increasing frequency
in the management of vascular problems, including
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

congenital heart lesions, such as branch pulmonary


artery stenosis, pulmonary vein stenosis, or coarctation of the aorta; traumatic arterial injuries; arterial
dissection; cerebral vascular abnormalities; renovascular disease; APLA syndrome; surgical stenosis; and
venous disease.561-573 Although stents can be successfully used in infants aged , 1 year, the small vessel
size increases the risk of thrombosis.574 There are no
studies assessing the role of anticoagulation or antiplatelet therapy to avoid stent occlusion in children.
Clinicians commonly administer UFH at the time of
stent insertion followed by aspirin therapy.
Recommendation
2.37. For children having endovascular stents
inserted, we suggest administration of UFH
perioperatively (Grade 2C).
2.38 Dilated Cardiomyopathy
The etiology of cardiomyopathy in children is quite
different from adults. Postviral and idiopathic cardiomyopathies occur in otherwise well children, whereas
dilated cardiomyopathy occurs frequently during the
end stage of muscular dystrophies. Thrombosis remains
a significant cause of morbidity and mortality.262,575,576
In a cross-sectional study of children awaiting cardiac
transplant, 31% had acute PE confirmed by ventilation/perfusion scan or angiography.577 A series of
66 patients with dilated cardiomyopathy reported a
prevalence of thrombosis of 14%.578
There are no studies of anticoagulant prophylaxis
in pediatric patients. However, based on adult studies
and the apparent risk of PE and stroke in children
with cardiomyopathy, primary prophylaxis with warfarin (target INR, 2.5; range, 2.0-3.0) often is used.579
Recommendation
2.38. For pediatric patients with cardiomyopathy, we suggest VKAs no later than their activation on a cardiac transplant waiting list (Grade 2C).
Underlying values and preferences: Parents who
place a high value on avoiding the inconvenience,
discomfort, and limitations of anticoagulant monitoring and a lower value on the uncertain reduction
in thrombotic complications are unlikely to choose
VKA therapy for their children who are eligible for
transplant.
2.39 Primary Pulmonary Hypertension
Relatively little evidence directly addresses the
role of anticoagulant therapy as primary prophylaxis
in children with pulmonary hypertension.580-582 However, on the basis of adult data and the basic pathophyswww.chestpubs.org

iology of the disease, clinicians commonly administer


anticoagulant prophylaxis in children with primary
pulmonary hypertension.580,583,584 The ACCP guidelines for medical management of primary pulmonary
hypertension in adults recommend routine anticoagulant prophylaxis with VKAs, although there is variation with respect to the target range recommended.
The guidelines acknowledge that some centers use a
target INR of 2.0 (range, 1.7-2.5), whereas others use
a target INR of 2.5 (range, 2.0-3.0). The ideal time to
commence anticoagulant therapy in children is uncertain; starting at the same time as vasodilator or other
medical therapy is common.585-587 Some authors have
suggested that the presence of reduced cardiac output or polycythemia is required to justify anticoagulant therapy.581
Recommendation
2.39. For children with primary pulmonary
hypertension, we suggest starting anticoagulation with VKAs at the same time as other medical
therapy (Grade 2C).
2.40-2.42 Biologic and Mechanical Prosthetic
Heart Valves
Biologic prosthetic heart valves may be surgically
placed in infants and children with congenital or
acquired heart disease when their innate tricuspid,
pulmonary valve, or both are not surgically repairable.588 Mechanical valves are preferred for mitral
and aortic replacement, given the catastrophic consequences of valve failure in these anatomic positions.589
Patients with biologic prosthetic heart valves usually
receive an antiplatelet agent. TE and bleeding events
are uncommon with this therapy.590-594
There is no direct evidence describing optimal thromboprophylaxis in children with bioprosthetic heart
valves. Recommendations, therefore, must rely on
indirect evidence from adults and the associated
recommendations.595
Mechanical prosthetic heart valves may be surgically placed in infants and children with congenital
or acquired heart disease when their innate valve is
not surgically repairable.596 Thrombotic complications
associated with mechanical prosthetic heart valves are
well described in adults. For this reason, clinicians
generally use VKAs to prevent complications, which
include TE, valve thrombosis, and ischemic stroke.
In children, optimal strategies for thromboprophylaxis for mechanical heart valves are less clear.
Studies in children typically consists of retrospective
case series, with many of the studies including
small numbers of infants and children, a spectrum of
age ranges, and varied valve positions and types.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e773S

Antithrombotic regimens described to prevent TE


complications range from no anticoagulation to the
use of antiplatelet agents or VKAs. The outcome
events reported include TE (valve thrombosis and
stroke), bleeding, and mortality.
The incidence of TE in children with mechanical
valves is reported to be as high as 68% per patient-year
in children who receive acetylsalicyclic acid (ASA)597
and 27% per patient-year for children who received
no drug therapy.597 Bleeding, when reported, is
extremely rare.203,594,598-603 When VKAs are prescribed,
the incidence of TE is reduced, but there is an
increased bleeding incidence.203,590,594,599,600,602-613 A series
of 32 children routinely treated with phenprocoumon collected over a 22-year period reported a
10-year freedom of 89.1% (1.2%/patient-year) from
any anticoagulation-related adverse event.614
There are few prospective studies and no RCTs in
children. Recommendations are therefore based on
the high-quality evidence supporting anticoagulant
thromboprophylaxis in adults and the available evidence in children.
Recommendations
2.40-2.42. For children with biologic or mechanical prosthetic heart valves, we recommend that
clinicians follow the relevant recommendations
from the adult population.595
2.43 Bacterial Endocarditis
Infective endocarditis in children most frequently
affects children with underlying congenital heart disease or previous cardiac surgery. One of the major
sequelae of infective endocarditis is embolic phenomenon and stroke.615-617 Embolic complications may occur
in as many as 30% of patients with infective endocarditis.618 There remains debate about the role of
urgent surgical intervention for infective endocarditis
in children.619,620 For those children managed conservatively, the presence or risk of emboli often leads to
the question of the role of anticoagulation. Emboli
are usually septic and may represent vegetations with
or without thrombin deposition.
There are no comparative data with respect to the
value of anticoagulation over and above antibiotics
alone, and data on this subject in adults is conflicting.
The role of anticoagulation must be considered on a
case-by-case basis, incorporating the assumed embolic
risks, potential need for surgery, and bleeding risks.
There appears to be a small role for thrombolysis or
potential benefit from antiplatelet agents.
2.44 Ventricular Assist Devices
Ventricular assist devices (VADs) are being used
more often in children with cardiac failure (congene774S

ital or acquired) as either bridge to transplantation or


to cardiac recovery.621-626 There are a variety of VADs
available, many specifically developed for pediatric
use.621
Studies of these devices in infants and children are
mainly retrospective case series with outcomes being
survival to transplant or to cardiac recovery.622,626-642
Reported survival to transplant or to cardiac recovery
ranges from 50%630 to 83%.628 There are no goodquality studies evaluating the safety and efficacy of
anticoagulant or antiplatelet therapy in children on
VAD support to reduce TE. There is no standardized antithrombotic regimen; however, on the basis
of adult data and the catastrophic consequences of
circuit occlusion or embolic complications, anticoagulant therapy in combination with antiplatelet therapy
seems preferable over no therapy.
Recommendation
2.44. For children with VADs, we suggest administration of UFH (Grade 2C). We suggest starting
UFH between 8 and 48 h following implantation (Grade 2C). In addition, we suggest antiplatelet therapy (either aspirin or aspirin and
dipyridamole) to commence within 72 h of VAD
placement (Grade 2C). For children with VAD,
once clinically stable, we suggest switching from
UFH to either LMWH or VKA (target INR, 3.0;
range, 2.5-3.5) until transplanted or weaned
from VAD (Grade 2C).
2.45-2.46 Primary Prophylaxis for Venous
Access Related to Hemodialysis
CVADs and arteriovenous fistulas are frequently
used to provide venous access for children during
hemodialysis.643 Hemodialysis is used more frequently
than peritoneal dialysis in children.644 Pediatric patients
who receive hemodialysis through a CVAD may be at
increased risk of CVAD-related DVT because of the
large-bore catheters used and the fluid shifts associated with intermittent dialysis. The average survival of CVADs used for hemodialysis is reported
to be , 1 year,643 whereas arteriovenous fistulas may
have as much as a 59% 5-year survival.645 In a small
historical cohort study of children with arteriovenous
fistulas, primary prophylaxis with LMWH was more
effective than aspirin, which in turn was more effective at preventing thrombosis than no treatment.646
Recommendations
2.45. For patients undergoing hemodialysis via
an arteriovenous fistula, we suggest routine use
of VKAs or LMWH as fistula thromboprophylaxis
as compared with no therapy (Grade 2C).
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

2.46. For patients undergoing hemodialysis via


CVAD, we suggest routine use of VKAs or LMWH
for thromboprophylaxis as compared with no
therapy (Grade 2C).
2.47 Use of UFH or LMWH During Hemodialysis
Intermittent hemodialysis traditionally requires procedural anticoagulation to avoid thrombosis within
the artificial circuit. A substantial amount of data
exists in adults with respect to the benefit of using
either UFH or LMWH to maintain circuit patency
during hemodialysis. Both UFH647,648 and LMWH649,650
have been reported as safe in children with uremia.
Citrate has also been reported to be safe and effective, especially in patients at higher risk of bleeding.651
Care must be taken with dosing and monitoring heparin prophylaxis in children receiving hemodialysis
because inadvertent systemic anticoagulation with clinical bleeding can occur.652
Recommendation
2.47. For children having hemodialysis, we suggest the use of UFH or LMWH during hemodialysis to maintain circuit patency independent
of type of vascular access (Grade 2C).
2.48-2.50 Kawasaki Disease
During the acute phase, Kawasaki disease may
cause medium-vessel and large-vessel arteritis, arterial aneurysms, valvulitis, and myocarditis. Kawasaki
disease is the leading cause of acquired heart disease
in children in North America. Of particular concern
are coronary artery aneurysms that may stenose or
thrombose. Coronary artery aneurysms or ectasia
develop in 15% to 25% of untreated children and
may lead to myocardial infarction, sudden death, or
chronic coronary arterial insufficiency.653
Treatment of Kawasaki disease in the acute phase
is directed at reducing inflammation in the coronary
artery wall and preventing coronary thrombosis, whereas
long-term therapy in individuals who develop coronary
aneurysms is aimed at preventing myocardial ischemia
or infarction.653 In patients with Kawasaki disease,
aspirin is initially given in high doses (80-100 mg/kg
per day during the acute phase for up to 14 days) as an
antiinflammatory agent and then in lower doses as an
antiplatelet agent (3-5 mg/kg per day for 6 to 8 weeks)
to prevent coronary aneurysm thrombosis and subsequent infarction (the major cause of death in patients
with Kawasaki disease).653 Because concomitant use
of ibuprofen or other nonsteroidal antiinflammatory
drugs may interfere with the effectiveness of aspirin,
these agents should be avoided.654
A large, multicenter RCT of high-dose IV g-globulin
plus aspirin compared with aspirin alone demonstrated
www.chestpubs.org

that coronary artery abnormalities were present in


14 of 79 (18%) children in the aspirin group compared with three of 79 (4%) children in the g-globulin
plus aspirin group (P 5 .005).655 Recent studies have
focused on trying to risk stratify patients at presentation into those who will only require IV immunoglobulin at 1 g/kg as a single dose656 vs those who will
not respond to the standard two doses of 1 g/kg and
require even more-aggressive therapy.657 Methylprednisolone in the acute phase has been shown not
to be beneficial in a recent well designed RCT.658
In a small study, patients who were treated with
abciximab demonstrated greater regression in aneurysm diameter at early follow-up than historical control patients who received standard therapy alone.211
These findings suggest that treatment with abciximab
may promote vascular remodeling and warrant further study.
Because no prospective data exist to guide clinicians in choosing an optimal regimen for the prevention of thrombosis in patients with Kawasaki disease
with coronary artery disease, recommendations are
based on the known pathophysiology, retrospective
case series in children with Kawasaki disease, and
extrapolation from experience in adults with coronary
disease.653 Therapeutic regimens used in patients
with Kawasaki disease depend on the severity of coronary involvement and include antiplatelet therapy
with aspirin, with or without clopidogrel or dipyridamole; anticoagulant therapy with VKAs or LMWH;
or a combination of anticoagulant and antiplatelet
therapy, usually VKAs plus aspirin.653 Long-term
combined VKA and aspirin is reported to be associated
with a 91% cardiac event-free outcome at 10 years
and a bleeding complication rate of 1.7% per year.659
LMWH has been reported to be as effective as warfarin and may lead to increased aneurysm regression
compared with warfarin. For long-term therapy, patients
often are converted to warfarin therapy.660
When a coronary aneurysm expands rapidly, the
risk of thrombosis is particularly high. For this reason, some experts advocate the use of UFH with aspirin.653 The most common antithrombotic regimen for
patients with giant aneurysms is low-dose aspirin
together with warfarin, maintaining an INR of 2.0 to
2.5.653,661 Some physicians substitute a therapeutic
dose of LMWH for warfarin.653
For giant aneurysms with acute thrombosis, thrombolysis or surgery is reported to be useful, but there
are no comparative data.265,659,662,663 Thrombolysis usually is given as a front-loaded protocol.661 Comprehensive guidelines have been published.664
Recommendations
2.48. For children with Kawasaki disease, we
recommend aspirin in high doses (80-100 mg/kg
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e775S

per day during the acute phase for up to 14 days)


as an antiinflammatory agent, then in lower
doses (1-5 mg/kg per day for 6 to 8 weeks) as an
antiplatelet agent (Grade 1B). For children with
Kawasaki disease, we recommend IV g-globulin
(2 g/kg, single dose) within 10 days of the onset
of symptoms (Grade 1A).
2.49. For children with moderate or giant coronary aneurysms following Kawasaki disease, we
suggest that warfarin in addition to low-dose
aspirin be given as primary thromboprophylaxis
(Grade 2C).
2.50. For children with Kawasaki disease who
have giant aneurysms and acute coronary artery
thrombosis, we suggest thrombolysis or acute
surgical intervention (Grade 2 C).
2.51 CSVT in Children
The estimated incidence of pediatric CSVT is
0.6 per 100,000 children per year with . 40% occurring in neonates as previously discussed.383,665 Radiographic diagnosis of CSVT requires imaging of the
thrombus within cerebral sinuses and veins because
nearly one-half of children have normal-appearing
brain parenchyma and the location and characteristics of venous infarction are very nonspecific.381,383,666,667
Imaging of the cerebral venous system is required
including magnetic resonance venography or CT
venography.379,666,668
Clinical outcomes after pediatric CSVT include
death in 9% to 29% and neurologic deficits, headaches, and seizure disorders in more than one-half
of survivors.383,668 Among neurologic deficits, cognitive and behavioral deficits are common, and motor
deficits are less common. In children, radiologic
recanalization as early as 2 weeks after the onset of
clinical symptoms has been reported.669 Predictors
of poor outcome include presentation with venous
infarcts or seizures383 and, for death, presentation
with coma.668 In the Canadian Pediatric Ischemic
Stroke Registry, nearly 25% of children showed an
increased severity of neurologic deficits developing over
time, reinforcing the need for long-term follow-up.
In addition, 13% of children with CSVT developed
recurrent cerebral or systemic thrombosis.383
Initial therapy for CSVT includes hydration, antibiotics or surgery for foci of cranial infection, anticonvulsants for seizures, and measures aimed at
decreasing intracranial pressure, with close monitoring for optic nerve compression.670 However, historically poor outcomes and recurrent thrombosis
provide the impetus for anticoagulant therapy.671
Clinical trials are lacking in pediatric CSVT. Four
randomized, placebo-controlled trials of heparin in
e776S

adults with CSVT support a benefit of heparin.672-676


Most guidelines recommend anticoagulation for adults
with CSVT, even in the presence of hemorrhage.677
Pooled outcome data from 150 cases of pediatric
CSVT published between 1980 and 1996 showed that
among 136 children who were not given anticoagulants,
the frequency of death was 16%, and the frequency
of poor neurologic outcome was 22% (combined poor
outcome, 36.5%). Among 14 treated children mortality
was 14%, and poor neurologic outcome occurred in
none (combined poor outcome, 14%).678 Sbire et al668
reported a trend toward better survival with no
cognitive sequelae with anticoagulation (OR, 3.64;
95% CI, 0.98-13.5); results failed to exclude either a
beneficial or a detrimental effect on the outcome of
death (OR, 0.29; 95% CI, 0.03-2.89).
Single-center and small multicenter series in
children110,378,383,666,668,679-687 have shown that IV UFH
and subcutaneous LMWH can be used safely in children. Hemorrhage is uncommon in patients treated
with anticoagulants in all series.
In a study combining patients from several European centers, nonadministration of antithrombotic
treatment in clinical risk situations and in children
with idiopathic CSVT (n 5 3) was significantly associated with higher risk of recurrence (P , .001).
The type of anticoagulation therapy administered (eg,
the use of UFH and warfarin or the application of
LMWH) did not influence thrombosis-free survival
(P 5 .54).685
Most recently, a large, single-center cohort reported
that 56 of 79 (71%) children with CSVT received
acute anticoagulation.393 Major hemorrhage occurred
in three children, two of whom had pretreatment intracranial hemorrhage. Bleeds were all nonfatal, and
clinical outcome was favorable in 50%, similar to the
remaining patients (53%), which is consistent with
data in adults with CSVT and hemorrhage that show
that the benefit of anticoagulation still outweighs the
risk.672,673,676 Early follow-up imaging demonstrated
thrombus propagation in seven of 19 (37%) children
without and three of 44 (7%) children with anticoagulation (RR, 3.1; 95% CI, 1.6-5.8).393 Propagation was
associated with new venous infarcts in 40% children
and moderate or severe clinical outcome (OR, 4.3;
95% CI, 1.0-19.4). The authors concluded that anticoagulation was safe and that nontreatment was associated with propagation in more than one-third of
children. The presence or absence of thrombophilia
should not affect decisions with regard to treatment
intensity or duration.688
The efficacy of thrombolysis in adults with CSVT
remains uncertain, although there are sufficient data
to conclude that it can be given safely.689 Evidence
regarding thrombolysis in children,690-693 mechanical
dissolution of clots or thrombectomy,694-696 and surgical
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

decompression697-699 is confined to case reports that


have reported apparent success in isolated cases or
small series of seriously ill patients, including children, usually in coma and with extensive thrombosis
of superficial and deep venous structures.690-692
Recommendation
2.51. For children with CSVT without significant ICH, we recommend anticoagulation initially with UFH or LMWH and subsequently
with LMWH or VKA for a minimum of 3 months
relative to no anticoagulation (Grade 1B). In children who after 3 months of therapy still experience occlusion of CSVT or ongoing symptoms,
we suggest administration of a further 3 months
of anticoagulation (Grade 2C). For children with
CSVT with significant hemorrhage, we suggest
initial anticoagulation as for children without hemorrhage or radiologic monitoring of the
thrombosis at 5 to 7 days and anticoagulation
if thrombus extension is noted at that time
(Grade 2C). In children with CSVT and potentially recurrent risk factors (eg, nephrotic
syndrome, asparaginase therapy), we suggest
prophylactic anticoagulation at times of risk
factor recurrence (Grade 2C). We suggest thrombolysis, thrombectomy, or surgical decompression only in children with severe CSVT in whom
there is no improvement with initial UFH therapy (Grade 2C).
2.52 AIS in Children
Reported incidence rates for AIS varies between
two and 13 per 100,000 children per year.700-702 Outcomes from childhood AIS include death in 3% to
5% and permanent cognitive or motor disability in
30% to 80%.671,703,704 Survival rates are significantly
better than in adults, and children with stroke who do
not die acutely will probably survive beyond middle
age, and the treatment of the resulting comorbidity
will be extremely expensive. The health burden of this
disease entity is thus very large.
Initial therapy in childhood AIS aims to limit
extension of occlusive thrombosis and early recurrent
thrombotic stroke. Subsequently, maintenance therapy aims to prevent longer-term recurrence.705 Mechanisms of stroke in children include cardiogenic and
large-vessel dissection-related embolism; cerebral
vasculopathy, which may be transient or progressive;
sickle cell disease; and in situ thrombosis. The conditions underlying these three mechanisms for stroke
differ markedly in children compared with adults and
notably exclude atherosclerosis. Frequently, chronic
diseases of childhood or acute illnesses, including
www.chestpubs.org

systemic infection and dehydration, underlie AIS.


However, up to 15% of children with AIS have no
apparent risk factors.
The results of adult stroke trials testing antithrombotic treatments cannot be directly extrapolated to
children because of different mechanisms for thrombus formation in adults with atherosclerosis. To date,
no RCTs of antithrombotic therapy have been conducted in children with stroke. Antiplatelet, anticoagulant, and other therapies in children with AIS are
selected based on the perceived mechanism for arterial thrombosis associated with the underlying risk
factors. Several cohort studies of children with AIS
have assessed safety and failure rates for antithrombotic agents679,706; however, the largest data set is from
the International Pediatric Stroke Study (IPSS) group.
This consortium involves . 30 centers worldwide and
collects data using standardized case report forms.
The IPSS has published data on children with AISs
occurring between 2003 and 2007.704 There were
661 children with AIS (640 with acute treatment data,
612 with morbidity data, and 643 with mortality data).
Acute therapy included anticoagulation alone in
171 (27%) patients, antiplatelet therapy alone in
177 (28%), antiplatelet and anticoagulation in 103
(16%), and no antithrombotic treatment in 189 (30%).
Subtypes associated with any use of anticoagulation
were dissection (OR, 14.09; 95% CI, 5.78-37.01) and
cardiac disease (OR, 1.87; 95% CI, 1.20-2.92). Factors associated with nonuse of anticoagulation included
sickle cell disease subtype (OR, 0.12; 95% CI, 0.020.95) and the enrollment center being located in the
United States (OR, 0.56; 95% CI, 0.39-0.80). Antiplatelet use was associated with Moyamoya (OR, 4.88;
95% CI, 2.13-11.12), whereas nonuse was associated
with dissection (OR, 0.47; 95% CI, 0.22-0.99), low
level of consciousness (OR, 0.45; 95% CI, 0.31-0.64),
and bilateral ischemia (OR, 0.32; 95% CI, 0.20-0.52).
Outcomes at hospital discharge included neurologic
deficits in 453 (74%) patients and death in 22 (3%). In
multivariate analysis, arteriopathy, bilateral ischemia,
and decreased consciousness at presentation were
prognostic of adverse outcome.704
When ASA therapy fails or is not tolerated in children with AIS, clopidogrel frequently is used. Risks
of combination therapy with ASA plus clopidogrel,
however, were recently highlighted by a study of
17 children who received clopidogrel (nine alone,
eight concurrent with aspirin) in whom two had subdural hemorrhages (both also receiving aspirin and
both having marked cerebral atrophy [1 Moyamoya,
1 progeria vasculopathy]).707
There are few data addressing the safety or efficacy
of tPA in children with AIS, and the literature associating outcomes with this treatment consists mostly
of isolated case reports.708,709 Although rarely feasible,
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e777S

older children with acute AIS may be diagnosed


within the time window for this treatment.
In Situ Thrombosis or Stroke of Undetermined Cause:
In situ thrombosis may be idiopathic; secondary to
local inflammation; or secondary to prothrombotic
conditions, including iron deficiency anemia,710-712
hyperhomocysteinemia,712-716 elevated levels of lipoprotein(a),218 and inherited prothrombotic disorders.218,717,718 The overall risk of a recurrent AIS and
TIA is 10% to 35%.413,415,719-729 The recurrence risk
increases in the presence of multiple stroke risk factors.726 Genetic thrombophilia was previously reported
as increasing the recurrence rate725; however, a more
recent comprehensive study suggested that this is
not the case.688 Recurrence risk is greatest in the
initial weeks and months following an index AIS
but persists for at least several years.413,725 Recurrent
stroke can be silent; infarction is documented in
one-third of children with cryptogenic stroke (not
due to obvious preexisting diseases) undergoing repeat
neuroimaging.725
Recommendation
2.52. For children with acute AIS with or without thrombophilia, we recommend UFH or
LMWH or aspirin as initial therapy until dissection and embolic causes have been excluded
(Grade 1C). For children with acute AIS, we suggest, once dissection and cardioembolic causes
are excluded, daily aspirin prophylaxis for a
minimum of 2 years as compared with no antithrombotic therapy (Grade 2C). For children
receiving aspirin who have recurrent AIS or
TIAs, we suggest changing to clopidogrel or
anticoagulant therapy with LMWH or VKA
(Grade 2C). For children with AIS, we recommend against the use of thrombolysis (tPA) or
mechanical thrombectomy outside of specific
research protocols (Grade 1C).
2.53 Embolic Stroke
Congenital heart disease and related interventions
(surgery or catheterization) are associated with paradoxical embolism through intracardiac defects. In the
setting of acute emolic stroke, the main principle of
treatment is to prevent further embolic phenomenon.
Strategies therefore target the source thrombosis,
even though this may not be visualized through standard imaging techniques. Thus, anticoagulation is the
preferred therapy because it is more effective than
antiplatelet therapy in the treatment of intracardiac
thrombosis and peripheral or central DVT.671 The
finding of a PFO on echocardiography in a child with
stroke without an associated documented venous
thrombosis often creates difficulties because the dise778S

tinction between an in situ thrombosis as a cause of


the AIS vs an embolic stroke may be impossible.730-732
Recommendation
2.53. For AIS secondary to cardioembolic causes,
we suggest anticoagulant therapy with LMWH
or VKAs for at least 3 months (Grade 2C). For
AIS secondary to cardioembolic causes in children with demonstrated right-to-left shunts (eg,
PFO), we suggest surgical closure of the shunt
(Grade 2C).
2.54 Dissection
Dissection of craniocervical arteries underlies 7%
of childhood AIS.733 In children with cerebral arterial
dissection underlying AIS, the risk of recurrent strokes
is 12%.733-735 Recurrence appears to be reduced
by antithrombotic treatment734 but is still observed
during anticoagulation733-735 or antiplatelet treatment.734
In adults with cerebral artery dissection, a Cochrane
meta-analysis that included 327 patients reported
no significant difference for initial or recurrent stroke
during anticoagulant treatment (five of 414) vs antiplatelet treatment (six of 157). The frequency of
major hemorrhage was 0.5% during anticoagulation.736 Subsequently, a large trial, the Spontaneous
vs Traumatic Arterial Dissection (SPONTADS) study,
showed recurrent stroke in two of 71 patients receiving anticoagulation treatment and one of 23 patients
receiving aspirin treatment.737 If data from the 105
SPONTADS patients are added to those pooled in
the Cochrane analysis, there is a trend showing benefit of anticoagulant therapy (seven of 485 stroke
on treatment) over antiplatelet therapy (seven of 180
stroke on treatment) (Fisher test P 5 .066; RR, 1.88;
95% CI, 1.10-3.23).
Recommendations
2.54. For AIS secondary to dissection, we suggest
anticoagulant therapy with LMWH or VKAs for
at least 6 weeks (Grade 2C). Ongoing treatment
will depend on radiologic assessment of degree
and extent of stenosis and evidence of recurrent
ischemic events.
2.55 Cerebral Vasculopathies
Cerebral vasculopathies can be inflammatory, traumatic, or idiopathic. Postvaricella angiopathy and
transient cerebral arteriopathy (or nonprogressive
primary angitis of the CNS) are among the most frequently seen and represent a unilateral inflammatory
process involving the intracranial vessels that comprise the circle of Willis.738 The recurrence rate for
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

vasculopathy may be increased compared with idiopathic stroke in children.413,725,739 Sequential imaging
studies may be required to differentiate the diagnosis, and ancillary studies (eg, varicella serology) are
important. Determination of the specific subtype of vasculopathy and monitoring of cerebral vessel appearance on magnetic resonance angiography or formal
angiography are critical for determining ongoing therapy requirements. In cerebral vasculitis, immunosuppressive agents may be required.740
Recommendation
2.55. For children with acute AIS secondary to
non-Moyamoya vasculopathy, we recommend
UFH or LMWH or aspirin for 3 months as initial
therapy compared with no treatment (Grade 1C).
For children with AIS secondary to nonMoyamoya vasculopathy, we suggest that ongoing
antithrombotic therapy should be guided by
repeat cerebrovascular imaging.
2.56-2.57 Moyamoya Disease
The most severe childhood cerebral vasculopathy
is Moyamoya, a progressive bilateral intracranial cerebral arteriopathy with severe stenosis or occlusion
of the terminal internal carotid arteries, typically
accompanied by basal collateral vessels.671 Recurrent
sequential infarcts, some silent, often are present at
diagnosis. The mechanisms for ischemia and infarction likely involved both chronic underperfusion and
thrombotic occlusion. Clinical presentations include
recurrent abrupt AIS and TIA presentations and progressive cognitive loss. Children with vascular stenosis or Moyamoya have a risk of recurrence as high
as 66%.413,725,741 Direct and indirect revascularization procedures to bypass the stenotic and occluded
arteries are available to increase regional cerebral
blood flow and reduce the risk of recurrence,742,743
and a meta-analysis of . 1,000 children confirmed
that surgery confers symptomatic benefit in almost
90% of children.744 A summary of evidence supporting the variety of surgical interventions is available.671
Anticoagulation is less frequently used because of
concerns about bleeding; however, the use of antiplatelet therapy is common. Few data confirm benefit in either the short or long term, although some
studies suggest that medical therapy is important perioperatively to reduce the risk of procedure-associated
strokes, which are common.745
Recommendations
2.56. For children with acute AIS secondary
to Moyamoya, we suggest aspirin over no treatment as initial therapy (Grade 2C).
www.chestpubs.org

2.57. For children with Moyamoya, we suggest


that they be referred to an appropriate center
for consideration of revascularization.
2.58-2.59 Sickle Cell Anemia
Children with sickle cell anemia can experience
stroke related to occlusion of small cerebral arteries
or through the development of Moyamoya. In sickle
cell anemia with stroke, reinfarction occurs in 7.06 of
100 patient-years despite treatment.746 In children
with sickle cell anemia receiving no treatment, recurrence is as high as 92%.747-755 Children with sickle
cell anemia and transcranial Doppler (TCD) velocities . 200 cm/s have a 40% risk of stroke over the
next 3 years.756 An RCT found significant reduction
in risk by blood transfusion every 6 weeks to decrease
hemoglobin S percentage to , 30%.757 Patients should
receive regular transfusions indefinitely as the risk of
overt stroke or reversion to high-risk TCD increases
when blood transfusions stop (STOP2 [Optimizing
Primary Stroke Prevention in Sickle Cell Anemia]).758
Hydroxyurea may also reduce stroke risk in children
with TCD velocities . 200 cm/s.759 Overt stroke is twice
as common in children with silent or covert infarction in the context of sickle cell anemia.746 Bone
marrow transplantation760-762 and revascularization for
Moyamoya763-766 are additional options for selected
patients; however, no RCTs have been completed for
these therapies. For specific recommendations related
to sickle cell disease, we refer the reader to the most
recent version of the more authoritative National Institutes of Health sickle cell treatment guidelines.767
Conclusion
The ninth edition of the antithrombotic therapy in
neonates and children guidelines presents 59 recommendations linked in a transparent manner to the
evidence on which the recommendations are based.
The guidelines address generic issues related to the
use of antithrombotic therapies in neonates and children as well as many specific clinical situations in
which these therapies are considered. Although there
has been considerable progress made in this field
over recent years, there remain many questions and
many gaps in our knowledge. However, there is no
doubt that the management of thrombosis in neonates and children is an increasing problem for clinicians, and it is hoped that these guidelines will provide
some degree of uniformity of approach while further
research is being undertaken.
Areas of Future Research
These guidelines highlight once again the lack
of evidence for many of the fundamental questions
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e779S

facing clinicians dealing with TE disease in neonates


and children. Immediate research priorities include
the following:
1. Natural history of disease: Many thromboses in
neonates, and indeed children, are now discovered incidentally or as part of routine screening.
Clear evidence that all thromboses in neonates
or children require treatment is lacking, and
studies assessing the long-term outcomes, particularly in asymptomatic thrombosis, are required
so that adequate risk-benefit assessments of
treatment options can be determined. In particular, uniform assessment criteria of complications such as PTS are required.
2. Intensity of anticoagulant therapy: The efficacy
and safety assessments of all anticoagulants used
in neonates and children remain uncertain partly
because there are no clinical outcome studies
documenting optimal therapeutic strategies (and
partly because there is as yet no uniform assessment of bleeding in the child treated with anticoagulation). Multiple publications now describe
the inadequacies of current therapeutic ranges
and of the monitoring tests used in current clinical practice. Therapeutic monitoring imposes a
considerable burden of care. No viable alternative strategy has been reported. The potential
for using weight-adjusted dosing without monitoring, particularly for UFH and LMWH, needs
to be explored.
3. Duration of therapy: Currently, duration of therapy for venous thrombosis in neonates and children is extrapolated from adult practice, despite
considerable evidence that this may not be relevant. Current clinical convention around duration
of therapy for many types of arterial thrombosis
seems entirely empirical. Multicenter clinical
outcome studies are required to address these
questions.
4. Role of nonpharmacologic interventions: Individual reports of interventional radiology or
surgical therapies for thrombosis in children are
increasing in frequency; however, there remain
no comparative studies and, in fact, few if any
consecutive cohort studies that enable patient
selection criteria to be adequately determined
or likely risk-benefit ratios of such interventions
to be considered.
5. Nonhematologic complications of therapy: Normal long-term physical, neurologic, and psychosocial development should be the goal of all
pediatric treatments. There is evidence that
interventions aimed at the coagulation system may
affect variable aspects of development, such
as neurocognitive outcome or bone density. A
e780S

greater understanding of the nonhematologic


impact of anticoagulation therapies is required
for holistic management of neonates and children with thrombosis.
There are many additional areas that require research
to improve the outcomes for neonates and children
with thrombosis, often at an individual disease level
(eg, the use of antiplatelet vs anticoagulation therapy
in a variety of clinical situations). However, the issues
noted in this article are fundamental to progressing
many other specific questions. As mentioned at the
beginning of the article, new agents are now being
specifically studied in children, and it is important
that all these aspects are considered in study design
and implementation, or else history will repeat itself,
and in years to come, we will regret missed opportunities to build a sustainable evidence base on which
to improve the care for neonates and children with
thrombosis.
Acknowledgments
Author contributions: As Topic Editor, Dr Vesely oversaw
the development of this article, including the data analysis and
subsequent development of the recommendations contained
herein.
Dr Monagle: contributed as Deputy Editor.
Dr Chan: contributed as a panelist.
Dr Goldenberg: contributed as a panelist.
Dr Ichord: contributed as a panelist.
Dr Journeycake: contributed as a panelist.
Dr Nowak-Gttl: contributed as a panelist.
Dr Vesely: contributed as Topic Editor.
Financial/nonfinancial disclosures: The authors of this guideline provided detailed conflict of interest information related
to each individual recommendation made in this article. A grid
of these disclosures is available online at http://chestjournal.
chestpubs.org/content/141/2_suppl/e737S/suppl/DC1. In summary, the
authors have reported to CHEST the following conflicts of
interest: Dr Goldenberg has received an NIH career development award and an investigator-initiated study grant from
Eisai Co., Ltd. He is also Chair of the steering committee for a
Phase II study of Dalteparin for Eisai Co. and Chair of a data monitoring committee for Bristol-Myers Squibb. Dr Ichord is a member of the Clinica Event Committee for Berlin Hearts IDE trial
of the EXCOR-Pediatric (pediatric ventricular assist device). This
involved reimbursement for travel expenses for study meetings
and for time spent on committee meetings for a total financial reimbursement or , $1,000/y from 2007 to 2012. Drs Monagle, Chan,
and Nowak-Gttl have contracted with Bayer (rivaroxaban).
Dr Journeycake has received honoraria from hemophilia companies (CSL, Baxter, Novo-Nordisk). Dr Vesely has reported that
no potential conflicts of interest exist with any companies/organizations whose products or services may be discussed in this article.
Role of sponsors: The sponsors played no role in the development of these guidelines. Sponsoring organizations cannot recommend panelists or topics, nor are they allowed prepublication
access to the manuscripts and recommendations. Guideline panel
members, including the chair, and members of the Health & Science Policy Committee are blinded to the funding sources. Further details on the Conflict of Interest Policy are available online
at http://chestnet.org.
Other contributions: We thank Eliza Mertyn for contributions
related to the article citations. She managed the EndNote database and referencing in the present article and posted pdf copies
of . 800 articles to http://box.net for the present authors use and
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

for authors of subsequent versions. We also thank Susan Millard,


MD, and Julie Zimbelman, MD, for their contributions.
Endorsements: This guideline is endorsed by the American
Association for Clinical Chemistry, the American College of Clinical Pharmacy, the American Society of Health-System Pharmacists, the American Society of Hematology, and the International
Society of Thrombosis and Hematosis.
Additional information: Table S1 can be found in the Online Supplement at http://chestjournal.chestpubs.org/content/141/2_suppl/
e737S/suppl/DC1.

References
1. Prandstetter C, Tamesberger M, Wagner O, et al. Medical
prescriptions to premature and newborn infants in an
Austrian neonatal intensive care unit [in German]. Klin
Padiatr. 2009;221(5):312-317.
2. Conroy S, Choonara I, Impicciatore P, et al; European
Network for Drug Investigation in Children. Survey of
unlicensed and off label drug use in paediatric wards in
European countries. BMJ. 2000;320(7227):79-82.
3. Monagle P, Chalmers E, Chan A, et al; American College of
Chest Physicians. Antithrombotic therapy in neonates and
children: American College of Chest Physicians EvidenceBased Clinical Practice Guidelines (8th Edition). Chest.
2008;133(suppl 6):887S-968S.
4. Monagle P, Ignjatovic V, Savoia H. Hemostasis in neonates and children: pitfalls and dilemmas. Blood Rev. 2010;
24(2):63-68.
5. Newall F, Ignjatovic V, Johnston L, et al. Age is a determinant factor for measures of concentration and effect in children requiring unfractionated heparin. Thromb Haemost.
2010;103(5):1085-1090.
6. European Medicines Agency. Guideline on Clinical
Investigation of Medicinal Products for Prophylaxis of
Venous Thromboembolic Risk in Non-Surgical Patients.
Committee for Medicinal Products for Human Use (CHMP).
London, England: European Medicines Agency; 2006.
7. Newall F, Johnston L, Ignjatovic V, Monagle P. Unfractionated heparin therapy in infants and children. Pediatrics.
2009;123(3):e510-e518.
8. MacLean S, Mulla S, Akl EA, et al. Patient values and preferences in decision making for antithrombotic therapy: a
systematic review: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest.
2012;141(2)(suppl):e1S-e23S.
9. Thomas DM, Albritton KH, Ferrari A. Adolescent and
young adult oncology: an emerging field. J Clin Oncol. 2010;
28(32):4781-4782.
10. Guyatt GH, Norris SL, Schulman S, et al. Methodology for
the development of antithrombotic therapy and prevention
of thrombosis guidelines: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest
Physicians evidence-based clinical practice guidelines. Chest.
2012;141(2)(suppl):53S-70S.
11. Hardikar W, Poddar U, Chamberlain J, et al. Evaluation of a
post-operative thrombin inhibitor replacement protocol to
reduce haemorrhagic and thrombotic complications after paediatric liver transplantation. Thromb Res. 2010;126(3):191-194.
12. Andrew M, Mitchell L, Vegh P, Ofosu F. Thrombin regulation in children differs from adults in the absence and presence of heparin. Thromb Haemost. 1994;72(6):836-842.
13. Gibson B, Chalmers E, Bolton-Maggs P, et al. Thromboembolism in childhood: a prospective two year BPSU study
in the United Kingdom. J Thromb Haemost. 2003;1(suppl 1):
OC422.
www.chestpubs.org

14. Kuhle S, Massicotte P, Chan A, et al. Systemic thromboembolism in children. Data from the 1-800-NO-CLOTS
Consultation Service. Thromb Haemost. 2004;92(4):722-728.
15. Massicotte P, Leaker M, Marzinotto V, et al. Enhanced
thrombin regulation during warfarin therapy in children compared to adults. Thromb Haemost. 1998;80(4):
570-574.
16. Mitchell LG, Andrew M, Hanna K, et al; Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic
Leukemia Treated with Asparaginase Group (PARKAA). A
prospective cohort study determining the prevalence of
thrombotic events in children with acute lymphoblastic
leukemia and a central venous line who are treated with
L-asparaginase: results of the Prophylactic Antithrombin
Replacement in Kids with Acute Lymphoblastic Leukemia
Treated with Asparaginase (PARKAA) Study. Cancer. 2003;
97(2):508-516.
17. Monagle P, Adams M, Mahoney M, et al. Outcome of
pediatric thromboembolic disease: a report from the Canadian Childhood Thrombophilia Registry. Pediatr Res. 2000;
47(6):763-766.
18. Newall F, Wallace T, Crock C, et al. Venous thromboembolic disease: a single-centre case series study. J Paediatr
Child Health. 2006;42(12):803-807.
19. Nowak-Gttl U, Heinecke A, von Kries R, Nrnberger W,
Mnchow N, Junker R. Thrombotic events revisited in
children with acute lymphoblastic leukemia: impact of concomitant Escherichia coli asparaginase/prednisone administration. Thromb Res. 2001;103(3):165-172.
20. Nowak-Gttl U, von Kries R, Gbel U. Neonatal symptomatic thromboembolism in Germany: two year survey. Arch
Dis Child Fetal Neonatal Ed. 1997;76(3):F163-F167.
21. Schmidt B, Andrew M. Neonatal thrombosis: report of a
prospective Canadian and international registry. Pediatrics.
1995;96(5 pt 1):939-943.
22. van Ommen CH, Heijboer H, Bller HR, Hirasing RA,
Heijmans HS, Peters M. Venous thromboembolism in childhood: a prospective two-year registry in The Netherlands.
J Pediatr. 2001;139(5):676-681.
23. White RH. The epidemiology of venous thromboembolism.
Circulation. 2003;107(23)(suppl 1):I-4-I-8.
24. Raffini L, Huang YS, Witmer C, Feudtner C. Dramatic
increase in venous thromboembolism in childrens hospitals in the United States from 2001 to 2007. Pediatrics.
2009;124(4):1001-1008.
25. Greenway A, Massicotte MP, Monagle P. Neonatal thrombosis and its treatment. Blood Rev. 2004;18(2):75-84.
26. Monagle P, Barnes C, Ignjatovic V, et al. Developmental
haemostasis. Impact for clinical haemostasis laboratories.
Thromb Haemost. 2006;95(2):362-372.
27. Ignjatovic V, Summerhayes R, Than J, Gan A, Monagle P.
Therapeutic range for unfractionated heparin therapy:
age-related differences in response in children. J Thromb
Haemost. 2006;4(10):2280-2282.
28. Newall F, Johnston L, Ignjatovic V, Summerhayes R,
Monagle P. Age-related plasma reference ranges for two
heparin-binding proteinsvitronectin and platelet factor 4.
Int J Lab Hematol. 2009;31(6):683-687.
29. Ignjatovic V, Straka E, Summerhayes R, Monagle P. Agespecific differences in binding of heparin to plasma proteins.
J Thromb Haemost. 2010;8(6):1290-1294.
30. Newall F, Barnes C, Ignjatovic V, Monagle P. Heparininduced thrombocytopenia in children. J Paediatr Child
Health. 2003;39(4):289-292.
31. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral
anticoagulants: antithrombotic therapy and prevention of
thrombosis, 9th ed: American College of Chest Physicians
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e781S

32.

33.
34.
35.
36.
37.
38.
39.

40.
41.

42.
43.
44.
45.

46.

47.

48.
49.
50.

51.

evidence-based clinical practice guidelines. Chest. 2012;


141(2)(suppl):e24S-e43S.
Andrew M, Schmidt B, Mitchell L, Paes B, Ofosu F.
Thrombin generation in newborn plasma is critically dependent on the concentration of prothrombin. Thromb Haemost.
1990;63(1):27-30.
Andrew M. The hemostatic system in the infant. In:
Nathan D, Oski F, eds. Hematology of Infancy and Childhood.
Philadelphia, PA: WB Saunders; 1992:115-154.
Andrew M, Vegh P, Johnston M, Bowker J, Ofosu F,
Mitchell L. Maturation of the hemostatic system during
childhood. Blood. 1992;80(8):1998-2005.
Ignjatovic V, Summerhayes R, Gan A, et al. Monitoring
unfractionated heparin (UFH) therapy: which anti-factor
Xa assay is appropriate? Thromb Res. 2007;120(3):347-351.
Newall F, Ignjatovic V, Summerhayes R, et al. In vivo age
dependency of unfractionated heparin in infants and children. Thromb Res. 2009;123(5):710-714.
Ignjatovic V, Furmedge J, Newall F, et al. Age-related differences in heparin response. Thromb Res. 2006;118(6):
741-745.
Hirsh J. Heparin. N Engl J Med. 1991;324(22):1565-1574.
Schmidt B, Gillie P, Mitchell L, Andrew M, Caco C,
Roberts R. A placebo-controlled randomized trial of antithrombin therapy in neonatal respiratory distress syndrome.
Am J Respir Crit Care Med. 1998;158(2):470-476.
Newall F, Ignjatovic V, Johnston L, et al. Clinical use of
unfractionated heparin therapy in children: time for change?
Br J Haematol. 2010;150(6):674-678.
Cuker A, Raby A, Moffat KA, Flynn G, Crowther MA.
Interlaboratory variation in heparin monitoring: lessons from
the Quality Management Program of Ontario coagulation
surveys. Thromb Haemost. 2010;104(4):837-844.
Kovacs MJ, Keeney M. Inter-assay and instrument variability
of anti-Xaresults. Thromb Haemost. 2000;84(1):138.
Mitchell LG, Vegh P. Conventional chromogenic heparin
assays are influenced by patients endogenous plasma antithrombin levels. Klin Padiatr. 2010;222(3):164-167.
Andrew M, Marzinotto V, Massicotte P, et al. Heparin
therapy in pediatric patients: a prospective cohort study.
Pediatr Res. 1994;35(1):78-83.
Raschke RA, Reilly BM, Guidry JR, Fontana JR, Srinivas S.
The weight-based heparin dosing nomogram compared
with a standard care nomogram. A randomized controlled
trial. Ann Intern Med. 1993;119(9):874-881.
Andrew M, Michelson AD, Bovill T, et al. The prevention
and treatment of thromboembolic disease in children: a
need for thrombophilia programs. J Pediatr Hematol Oncol.
1997;19(1):7-22.
Schroeder AR, Axelrod DM, Silverman NH, Rubesova E,
Merkel E, Roth SJ. A continuous heparin infusion does not
prevent catheter-related thrombosis in infants after cardiac
surgery. Pediatr Crit Care Med. 2010;11(4):489-495.
McDonald MM, Jacobson LJ, Hay WW Jr, Hathaway WE.
Heparin clearance in the newborn. Pediatr Res. 1981;15(7):
1015-1018.
Turner Gomes S, Nitschmann E, Benson L, et al. Heparin is
cleared faster in children with congenital heart disease than
adults [abstract]. J Am Coll Cardiol. 1993;21:59a.
Kuhle S, Eulmesekian P, Kavanagh B, et al. Lack of correlation between heparin dose and standard clinical monitoring
tests in treatment with unfractionated heparin in critically ill
children. Haematologica. 2007;92(4):554-557.
Ho SH, Wu JK, Hamilton DP, Dix DB, Wadsworth LD.
An assessment of published pediatric dosage guidelines for
enoxaparin: a retrospective review. J Pediatr Hematol Oncol.
2004;26(9):561-566.

e782S

52. Trame MN, Mitchell L, Krmpel A, Male C, Hempel G,


Nowak-Gttl U. Population pharmacokinetics of enoxaparin in infants, children and adolescents during secondary
thromboembolic prophylaxis: a cohort study. J Thromb
Haemost. 2010;8(9):1950-1958.
53. Michelson AD, Bovill E, Andrew M. Antithrombotic therapy
in children. Chest. 1995;108(4 suppl):506S-522S.
54. Kuhle S, Eulmesekian P, Kavanagh B, Massicotte P, Vegh P,
Mitchell LG. A clinically significant incidence of bleeding in
critically ill children receiving therapeutic doses of unfractionated heparin: a prospective cohort study. Haematologica.
2007;92(2):244-247.
55. Galant SP. Accidental heparinization of a newborn infant.
Am J Dis Child. 1967;114(3):313-319.
56. Glueck HI, Light IJ, Flessa H, Sutherland JM. Inadvertent
sodium heparin administration to a newborn infant. JAMA.
1965;191:1031-1032.
57. Moncino MD, Kurtzberg J. Accidental heparinization in the
newborn: a case report and brief review of the literature.
J Perinatol. 1990;10(4):399-402.
58. Pachman DJ. Accidental heparin poisoning in an infant.
Am J Dis Child. 1965;110:210-212.
59. Pegelow CH, Powars D. Inadvertent heperanization as a
complication of intensive care [abstract]. Clin Res. 1975;23:
161A.
60. Schreiner RL, Wynn RJ, McNulty C. Accidental heparin
toxicity in the newborn intensive care unit. J Pediatr. 1978;
92(1):115-116.
61. Murphy MS, John PR, Mayer AD, Buckels JA, Kelly DA.
Heparin therapy and bone fractures. Lancet. 1992;340(8827):
1098.
62. Sackler JP, Liu L. Heparin-induced osteoporosis. Br J
Radiol. 1973;46(547):548-550.
63. Rauch F, Travers R, Plotkin H, Glorieux FH. The effects
of intravenous pamidronate on the bone tissue of children
and adolescents with osteogenesis imperfecta. J Clin Invest.
2002;110(9):1293-1299.
64. Shaughnessy SG, Young E, Deschamps P, Hirsh J. The
effects of low molecular weight and standard heparin
on calcium loss from fetal rat calvaria. Blood. 1995;86(4):
1368-1373.
65. Klenner A, Greinacher A. Heparin-induced thrombocytopenia in children. In: Warkentin T, Greinacher A, eds.
Heparin-Induced Thrombocytopenia. 3rd ed. New York,
NY: Marcel Dekker Inc; 2004:553-571.
66. Boshkov L, Ibsen L, Kirby A, Ungerleider R, Shen I. Heparininduced thrombocytopenia (HIT) in neonates and very young
children undergoing congenital cardiac surgery: a likely
under-recognized complication with significant morbidity
and mortality: report of 4 sequential cases [abstract]. J Thromb
Haemost. 2003;1(suppl 1):1494a.
67. Etches W, Stang L, Conradi A. Incidence of heparininduced thrombocytopenia in a pediatric intensive care population [abstract]. Blood. 2003;102(suppl):536a.
68. Schmugge M, Risch L, Huber AR, Benn A, Fischer JE.
Heparin-induced thrombocytopenia-associated thrombosis in
pediatric intensive care patients. Pediatrics. 2002;109(1):E10.
69. Spadone D, Clark F, James E, Laster J, Hoch J, Silver D.
Heparin-induced thrombocytopenia in the newborn. J Vasc
Surg. 1992;15(2):306-312.
70. Klenner AF, Lubenow N, Raschke R, Greinacher A.
Heparin-induced thrombocytopenia in children: 12 new
cases and review of the literature. Thromb Haemost. 2004;
91(4):719-724.
71. Chan VH, Monagle P, Massicotte P, Chan AK. Novel paediatric anticoagulants: a review of the current literature. Blood
Coagul Fibrinolysis. 2010;21(2):144-151.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

72. Murdoch IA, Beattie RM, Silver DM. Heparin-induced


thrombocytopenia in children. Acta Paediatr. 1993;82(5):
495-497.
73. Potter C, Gill JC, Scott JP, McFarland JG. Heparininduced thrombocytopenia in a child. J Pediatr. 1992;121(1):
135-138.
74. Risch L, Fischer JE, Herklotz R, Huber AR. Heparininduced thrombocytopenia in paediatrics: clinical characteristics, therapy and outcomes. Intensive Care Med. 2004;
30(8):1615-1624.
75. Saxon BR, Black MD, Edgell D, Noel D, Leaker MT.
Pediatric heparin-induced thrombocytopenia: management
with Danaparoid (orgaran). Ann Thorac Surg. 1999;68(3):
1076-1078.
76. Severin T, Sutor AH. Heparin-induced thrombocytopenia
in pediatrics. Semin Thromb Hemost. 2001;27(3):293-299.
77. Severin T, Zieger B, Sutor AH. Anticoagulation with recombinant hirudin and danaparoid sodium in pediatric patients.
Semin Thromb Hemost. 2002;28(5):447-454.
78. Almond CS, Harrington J, Thiagarajan R, et al. Successful
use of bivalirudin for cardiac transplantation in a child
with heparin-induced thrombocytopenia. J Heart Lung
Transplant. 2006;25(11):1376-1379.
79. Alsoufi B, Boshkov LK, Kirby A, et al. Heparin-induced
thrombocytopenia (HIT) in pediatric cardiac surgery: an
emerging cause of morbidity and mortality. Semin Thorac
Cardiovasc Surg Pediatr Card Surg Annu. 2004;7:155-171.
80. Bidlingmaier C, Magnani HN, Girisch M, Kurnik K. Safety
and efficacy of danaparoid (Orgaran) use in children. Acta
Haematol. 2006;115(3-4):237-247.
81. Cetta F, Graham LC, Wrona LL, Arruda MJ, Walenga JM.
Argatroban use during pediatric interventional cardiac catheterization. Catheter Cardiovasc Interv. 2004;61(1):147-149.
82. Dager WE, Gosselin RC, Yoshikawa R, Owings JT. Lepirudin
in heparin-induced thrombocytopenia and extracorporeal
membranous oxygenation. Ann Pharmacother. 2004;38(4):
598-601.
83. Deitcher SR, Topoulos AP, Bartholomew JR, KichukChrisant MR. Lepirudin anticoagulation for heparininduced thrombocytopenia. J Pediatr. 2002;140(2):264-266.
84. Dyke PC II, Russo P, Mureebe L, Russo J, Tobias JD.
Argatroban for anticoagulation during cardiopulmonary bypass
in an infant. Paediatr Anaesth. 2005;15(4):328-333.
85. Hassell K. Heparin-induced thrombocytopenia: diagnosis
and management. Thromb Res. 2008;123(suppl 1):S16-S21.
86. Hursting MJ, Dubb J, Verme-Gibboney CN. Argatroban
anticoagulation in pediatric patients: a literature analysis.
J Pediatr Hematol Oncol. 2006;28(1):4-10.
87. Iannoli ED, Eaton MP, Shapiro JR. Bidirectional Glenn
shunt surgery using lepirudin anticoagulation in an infant
with heparin-induced thrombocytopenia with thrombosis.
Anesth Analg. 2005;101(1):74-76.
88. John TE, Hallisey RK Jr. Argatroban and lepirudin requirements in a 6-year-old patient with heparin-induced thrombocytopenia. Pharmacotherapy. 2005;25(10):1383-1388.
89. Knoderer CA, Knoderer HM, Turrentine MW, Kumar M.
Lepirudin anticoagulation for heparin-induced thrombocytopenia after cardiac surgery in a pediatric patient.
Pharmacotherapy. 2006;26(5):709-712.
90. Mejak B, Giacomuzzi C, Heller E, et al. Argatroban usage
for anticoagulation for ECMO on a post-cardiac patient with
heparin-induced thrombocytopenia. J Extra Corpor Technol.
2004;36(2):178-181.
91. Napolitano LM, Warkentin TE, Almahameed A, Nasraway SA.
Heparin-induced thrombocytopenia in the critical care setting: diagnosis and management. Crit Care Med. 2006;34(12):
2898-2911.
www.chestpubs.org

92. Neuhaus TJ, Goetschel P, Schmugge M, Leumann E.


Heparin-induced thrombocytopenia type II on hemodialysis:
switch to danaparoid. Pediatr Nephrol. 2000;14(8-9):713-716.
93. Nguyen TN, Gal P, Ransom JL, Carlos R. Lepirudin use
in a neonate with heparin-induced thrombocytopenia. Ann
Pharmacother. 2003;37(2):229-233.
94. Nowak G. New anticoagulants for secondary haemostasis
anti IIa inhibitors [in German]. Hamostaseologie. 2009;29(3):
256-259.
95. Potter KE, Raj A, Sullivan JE. Argatroban for anticoagulation in pediatric patients with heparin-induced thrombocytopenia requiring extracorporeal life support. J Pediatr
Hematol Oncol. 2007;29(4):265-268.
96. Ranze O, Ranze P, Magnani HN, Greinacher A. Heparininduced thrombocytopenia in paediatric patientsa review
of the literature and a new case treated with danaparoid
sodium. Eur J Pediatr. 1999;158(suppl 3):S130-S133.
97. Scott LK, Grier LR, Conrad SA. Heparin-induced thrombocytopenia in a pediatric patient receiving extracorporeal
support and treated with argatroban. Pediatr Crit Care Med.
2006;7(3):255-257.
98. Warkentin TE. Think of HIT. Hematology (Am Soc Hematol
Educ Program). 2006;1:408-414.
99. Warkentin TE, Greinacher A, Koster A, Lincoff AM;
American College of Chest Physicians. Treatment and prevention of heparin-induced thrombocytopenia: American
College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) [published correction appears
in Chest. 2011 May;139(5):1261. Dosage error in article text].
Chest. 2008;133(6 suppl):340S-380S.
100. Dix D, Andrew M, Marzinotto V, et al. The use of low
molecular weight heparin in pediatric patients: a prospective cohort study. J Pediatr. 2000;136(4):439-445.
101. Merkel N, Gunther G, Schobess R. Long-term treatment
of thrombosis with enoxaparin in pediatric and adolescent
patients. Acta Haematol. 2006;115(3-4):230-236.
102. Ignjatovic V, Najid S, Newall F, Summerhayes R, Monagle P.
Dosing and monitoring of enoxaparin (Low molecular weight
heparin) therapy in children. Br J Haematol. 2010;149(5):
734-738.
103. Bontadelli J, Moeller A, Schmugge M, et al. Use of enoxaparin in the treatment of catheter-related arterial thrombosis in infants with congenital heart disease. Cardiol Young.
2006;16:27.
104. Burak CR, Bowen MD, Barron TF. The use of enoxaparin
in children with acute, nonhemorrhagic ischemic stroke.
Pediatr Neurol. 2003;29(4):295-298.
105. Dix D, Charpentier K, Sparling C, Massicotte MP. Determination of trough anti-factor Xa levels in pediatric patients
on low molecular weight heparin (LMWH). J Pediatr Hematol
Oncol. 1998;20(4):398, Abstract #667.
106. Dunaway KK, Gal P, Ransom JL. Use of enoxaparin in a preterm infant. Ann Pharmacother. 2000;34(12):1410-1413.
107. Elhasid R, Lanir N, Sharon R, et al. Prophylactic therapy
with enoxaparin during L-asparaginase treatment in children
with acute lymphoblastic leukemia. Blood Coagul Fibrinolysis.
2001;12(5):367-370.
108. Nowak-Gttl U, Bidlingmaier C, Krmpel A, Gttl L,
Kenet G. Pharmacokinetics, efficacy, and safety of LMWHs
in venous thrombosis and stroke in neonates, infants and
children. Br J Pharmacol. 2008;153(6):1120-1127.
109. Revel-Vilk S, Sharathkumar A, Massicotte P, et al. Natural
history of arterial and venous thrombosis in children treated
with low molecular weight heparin: a longitudinal study by
ultrasound. J Thromb Haemost. 2004;2(1):42-46.
110. Schobess R, Dring C, Bidlingmaier C, Heinecke A,
Merkel N, Nowak-Gttl U. Long-term safety and efficacy
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e783S

111.

112.

113.

114.

115.

116.

117.

118.

119.
120.

121.
122.
123.

124.

125.

126.

data on childhood venous thrombosis treated with a low


molecular weight heparin: an open-label pilot study of
once-daily versus twice-daily enoxaparin administration.
Haematologica. 2006;91(12):1701-1704.
Streif W, Goebel G, Chan AK, Massicotte MP. Use of low
molecular mass heparin (enoxaparin) in newborn infants:
a prospective cohort study of 62 patients. Arch Dis Child
Fetal Neonatal Ed. 2003;88(5):F365-F370.
Blatn J, Fiamoli V. Treatment of deep vein thrombosis with
continuous intravenous infusion of LMWH in children
an alternative to subcutaneous application when needed.
Vnitr Lek. 2009;55(3):227-232.
Massicotte P, Julian JA, Gent M, et al; PROTEKT Study
Group. An open-label randomized controlled trial of low
molecular weight heparin for the prevention of central
venous line-related thrombotic complications in children:
the PROTEKT trial. Thromb Res. 2003;109(2-3):101-108.
Kuhle S, Massicotte P, Dinyari M, et al. Dose-finding and
pharmacokinetics of therapeutic doses of tinzaparin in
pediatric patients with thromboembolic events. Thromb
Haemost. 2005;94(6):1164-1171.
Massicotte MP, Adams M, Leaker M, et al. A nomogram
to establish therapeutic levels of the low molecular weight
heparin (LMWH), clivarine in children requiring treatment
for venous thromboembolism (VTE). Thromb Haemost.
1997;(suppl):282.
Massicotte P, Julian JA, Marzinotto V, et al. Dose-finding
and pharmacokinetic profiles of prophylactic doses of a low
molecular weight heparin (reviparin-sodium) in pediatric
patients. Thromb Res. 2003;109(2-3):93-99.
Nohe N, Flemmer A, Rmler R, Praun M, Auberger K. The
low molecular weight heparin dalteparin for prophylaxis and
therapy of thrombosis in childhood: a report on 48 cases.
Eur J Pediatr. 1999;158(suppl 3):S134-S139.
Punzalan RC, Hillery CA, Montgomery RR, Scott CA,
Gill JC. Low-molecular-weight heparin in thrombotic disease in children and adolescents. J Pediatr Hematol Oncol.
2000;22(2):137-142.
Hofmann S, Knoefler R, Lorenz N, et al. Clinical experiences with low-molecular weight heparins in pediatric
patients. Thromb Res. 2001;103:345-353.
Massicotte P, Adams M, Marzinotto V, Brooker LA,
Andrew M. Low-molecular-weight heparin in pediatric patients with thrombotic disease: a dose finding study.
J Pediatr. 1996;128(3):313-318.
Vieira A, Berry L, Ofosu F, Andrew M. Heparin sensitivity
and resistance in the neonate: an explanation. Thromb Res.
1991;63(1):85-98.
van Ommen CH, van den Dool EJ, Peters M. Nadroparin
therapy in pediatric patients with venous thromboembolic
disease. J Pediatr Hematol Oncol. 2008;30(3):230-234.
Malowany JI, Monagle P, Knoppert DC, et al; Canadian
Paediatric Thrombosis and Hemostasis Network. Enoxaparin
for neonatal thrombosis: a call for a higher dose for neonates. Thromb Res. 2008;122(6):826-830.
Malowany JI, Knoppert DC, Chan AK, Pepelassis D,
Lee DS. Enoxaparin use in the neonatal intensive care
unit: experience over 8 years. Pharmacotherapy. 2007;27(9):
1263-1271.
Michaels LA, Gurian M, Hegyi T, Drachtman RA. Low
molecular weight heparin in the treatment of venous and
arterial thromboses in the premature infant. Pediatrics.
2004;114(3):703-707.
Crowther MA, Berry LR, Monagle PT, Chan AK. Mechanisms responsible for the failure of protamine to inactivate low-molecular-weight heparin. Br J Haematol. 2002;
116(1):178-186.

e784S

127. Harenberg J, Wrzner B, Zimmermann R, Schettler G.


Bioavailability and antagonization of the low molecular
weight heparin CY 216 in man. Thromb Res. 1986;44(4):
549-554.
128. Massonnet-Castel S, Pelissier E, Bara L, et al. Partial reversal of low molecular weight heparin (PK 10169) anti-Xa
activity by protamine sulfate: in vitro and in vivo study during
cardiac surgery with extracorporeal circulation. Haemostasis.
1986;16(2):139-146.
129. Thomas DP. Does low molecular weight heparin cause less
bleeding? Thromb Haemost. 1997;78(6):1422-1425.
130. Van Ryn-McKenna J, Cai L, Ofosu FA, Hirsh J, Buchanan MR.
Neutralization of enoxaparine-induced bleeding by protamine sulfate. Thromb Haemost. 1990;63(2):271-274.
131. Haroon Y, Shearer MJ, Rahim S, Gunn WG, McEnery G,
Barkhan P. The content of phylloquinone (vitamin K1) in
human milk, cows milk and infant formula foods determined by high-performance liquid chromatography. J Nutr.
1982;112(6):1105-1117.
132. von Kries R, Shearer M, McCarthy PT, Haug M, Harzer G,
Gbel U. Vitamin K1 content of maternal milk: influence of the
stage of lactation, lipid composition, and vitamin K1 supplements given to the mother. Pediatr Res. 1987;22(5):513-517.
133. Bonduel MM. Oral anticoagulation therapy in children.
Thromb Res. 2006;118(1):85-94.
134. Andrew M, Marzinotto V, Brooker LA, et al. Oral anticoagulation therapy in pediatric patients: a prospective study.
Thromb Haemost. 1994;71(3):265-269.
135. Carpentieri U, Nghiem QX, Harris LC. Clinical experience with an oral anticoagulant in children. Arch Dis Child.
1976;51(6):445-448.
136. Doyle JJ, Koren G, Cheng MY, Blanchette VS. Anticoagulation with sodium warfarin in children: effect of a loading
regimen. J Pediatr. 1988;113(6):1095-1097.
137. Evans DI, Rowlands M, Poller L. Survey of oral anticoagulant treatment in children. J Clin Pathol. 1992;45(8):
707-708.
138. Streif W, Andrew M, Marzinotto V, et al. Analysis of warfarin therapy in pediatric patients: A prospective cohort study
of 319 patients. Blood. 1999;94(9):3007-3014.
139. Bonduel M, Sciuccati G, Hepner M, et al. Acenocoumarol
therapy in pediatric patients. J Thromb Haemost. 2003;
1(8):1740-1743.
140. Piquet P, Losay J, Doubine S. Acenocoumarol (Sintrom)
and fluinidione (Previscan) in pediatrics after cardiac surgical
procedures [in French]. Arch Pediatr. 2002;9(11):1137-1144.
141. Newall F, Savoia H, Campbell J, Monagle P. Anticoagulation
clinics for children achieve improved warfarin management.
Thromb Res. 2004;114(1):5-9.
142. Bauman ME, Black KL, Massicotte MP, et al. Accuracy of
the CoaguChek XS for point-of-care international normalized ratio (INR) measurement in children requiring warfarin. Thromb Haemost. 2008;99(6):1097-1103.
143. Greenway A, Ignjatovic V, Summerhayes R, et al. Pointof-care monitoring of oral anticoagulation therapy in children. Comparison of the CoaguChek XS system with venous
INR and venous INR using an International Reference
Thromboplastin preparation (rTF/95). Thromb Haemost.
2009;102(1):159-165.
144. Marzinotto V, Monagle P, Chan A, et al. Capillary whole
blood monitoring of oral anticoagulants in children in outpatient clinics and the home setting. Pediatr Cardiol. 2000;
21(4):347-352.
145. Massicotte P, Marzinotto V, Vegh P, Adams M, Andrew M.
Home monitoring of warfarin therapy in children with a
whole blood prothrombin time monitor. J Pediatr. 1995;
127(3):389-394.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

146. Newall F, Monagle P, Johnston L. Home INR monitoring of


oral anticoagulant therapy in children using the CoaguChek
S point-of-care monitor and a robust education program.
Thromb Res. 2006;118(5):587-593.
147. Nowatzke WL, Landt M, Smith C, Wilhite T, Canter C,
Luchtman-Jones L. Whole blood international normalization
ratio measurements in children using near-patient monitors.
J Pediatr Hematol Oncol. 2003;25(1):33-37.
148. Bhat D, Upponi A, Rakecha A, Thomson J. Evaluating
safety, effectiveness, and user satisfaction of home international normalized ratio monitoring service: experience from
a tertiary pediatric cardiology unit in the United Kingdom.
Pediatr Cardiol. 2010;31(1):18-21.
149. Bradbury MJ, Taylor G, Short P, Williams MD. A comparative study of anticoagulant control in patients on long-term
warfarin using home and hospital monitoring of the international normalised ratio. Arch Dis Child. 2008;93(4):303-306.
150. Monagle P, Michelson AD, Bovill E, Andrew M. Antithrombotic therapy in children. Chest. 2001;119(1 suppl):
344S-370S.
151. Massicotte P, Julian JA, Gent M, et al; REVIVE Study Group.
An open-label randomized controlled trial of low molecular
weight heparin compared to heparin and coumadin for the
treatment of venous thromboembolic events in children: the
REVIVE trial. Thromb Res. 2003;109(2-3):85-92.
152. Newall F, Campbell J, Savoia H, et al. Incidence of major
bleeding in a large paediatric cohort of patients requiring
warfarin therapy. J Thromb Haemost. 2005;3(Supplement 1):
OR357.
153. Bauman ME, Black K, Kuhle S, et al; Western Canadian
Childrens Heart Network WCCHN. KIDCLOT: the importance of validated educational intervention for optimal
long term warfarin management in children. Thromb Res.
2009;123(5):707-709.
154. Newall F, Johnston L, Monagle P. Optimising anticoagulant
education in the paediatric setting using a validated model
of education. Patient Educ Couns. 2008;73(2):384-388.
155. Ries M, Klinge J, Rauch R. Erfahrungen mit der antikoagulatientherapie bei 10 patienten an der Univ Kinderklinik
Erlangen. In: Sutor AH, ed. Thrombosen im Kindersalter.
Basel, Switzerland: Editiones Roche; 1992.
156. Taybi H, Capitanio MA. Tracheobronchial calcification: an
observation in three children after mitral valve replacement and warfarin sodium therapy. Radiology. 1990;176(3):
728-730.
157. Barnes C, Newall F, Ignjatovic V, et al. Reduced bone density in children on long-term warfarin. Pediatr Res. 2005;
57(4):578-581.
158. Massicotte P, Julian J, Webber C. Osteoporosis: a potential complication of long term warfarin therapy [abstract].
Thromb Haemost. 1999;(suppl):1333a.
159. Bolton-Maggs P, Brook L. The use of vitamin K for reversal of over-warfarinization in children. Br J Haematol. 2002;
118(3):924.
160. Andrew M. Developmental hemostasis: relevance to thromboembolic complications in pediatric patients. Thromb
Haemost. 1995;74(1):415-425.
161. Bates SM, Weitz JI. The status of new anticoagulants. Br J
Haematol. 2006;134(1):3-19.
162. Young G, Tarantino MD, Wohrley J, Weber LC, Belvedere M,
Nugent DJ. Pilot dose-finding and safety study of bivalirudin in infants ,6 months of age with thrombosis. J Thromb
Haemost. 2007;5(8):1654-1659.
163. Bning A, Morschheuser T, Blse U, et al. Incidence of
heparin-induced thrombocytopenia and therapeutic strategies in pediatric cardiac surgery. Ann Thorac Surg. 2005;
79(1):62-65.
www.chestpubs.org

164. Dager WE, White RH. Low-molecular-weight heparininduced thrombocytopenia in a child. Ann Pharmacother.
2004;38(2):247-250.
165. Schuepbach RA, Meili EO, Schneider E, Peter U, Bachli EB.
Lepirudin therapy for thrombotic complications in congenital afibrinogenaemia. Thromb Haemost. 2004;91(5):
1044-1046.
166. Eikelboom JE, Weitz JI. Dabigatran etexilate for prevention of venous thromboembolism. Thromb Haemost. 2009;
101(1):2-4.
167. Zikria JC, Ansell J. Oral anticoagulation with factor Xa and
thrombin inhibitors: on the threshold of change. Curr Opin
Hematol. 2009;16(5):347-356.
168. Acostamadiedo JM, Iyer UG, Owen J. Danaparoid sodium.
Expert Opin Pharmacother. 2000;1(4):803-814.
169. Wilhelm MJ, Schmid C, Kececioglu D, Mllhoff T,
Ostermann H, Scheld HH. Cardiopulmonary bypass in
patients with heparin-induced thrombocytopenia using Org
10172. Ann Thorac Surg. 1996;61(3):920-924.
170. Zhrer B, Zenz W, Rettenbacher A, et al. Danaparoid
sodium (Orgaran) in four children with heparin-induced thrombocytopenia type II. Acta Paediatr. 2001;90(7):765-771.
171. Klement D , Rammos S , v Kries R , Kirschke W ,
Kniemeyer HW, Greinacher A. Heparin as a cause of thrombus progression. Heparin-associated thrombocytopenia is an
important differential diagnosis in paediatric patients even
with normal platelet counts. Eur J Pediatr. 1996;155(1):11-14.
172. Warkentin TE, Levine MN, Hirsh J, et al. Heparin-induced
thrombocytopenia in patients treated with low-molecularweight heparin or unfractionated heparin. N Engl J Med.
1995;332(20):1330-1335.
173. Weigel B, Lasky A, Krishnamurti L, et al. Danaparoid
(Orgaran) anticoagulation of pediatric patients with heparinDanaparoid (Orgaran) anticoagulation of pediatric patients
with heparin induced thrombocytopenia (HIT) [abstract].
J Pediatr Hematol Oncol. 1999;21(4):327.
174. Schiffmann H, Untrhalt M, Harms K, et al. Successful treatment of heparin-induced thrombocytopenia type II in childhood with recombinant hirudin [in German]. Monatsschr
Kinderheilkd. 1997;145(6):606-612.
175. Girisch M, Buheitel G, Ries M, et al. Safe and effective use
of danaparoid during cardiac catheterization in a 14 month
old boy with tetralogy of Fallot and heparin-induced thrombocytopenia [abstract]. Ann Hematol. 2001;80(suppl 1):A20.
176. Sharathkumar AA, Crandall C, Lin JJ, Pipe S. Treatment
of thrombosis with fondaparinux (Arixtra) in a patient with
end-stage renal disease receiving hemodialysis therapy.
J Pediatr Hematol Oncol. 2007;29(8):581-584.
177. Boshkov LK, Kirby A, Shen I, Ungerleider RM. Recognition
and management of heparin-induced thrombocytopenia in
pediatric cardiopulmonary bypass patients. Ann Thorac
Surg. 2006;81(6):S2355-S2359.
178. Young G, Nugent DJ. Use of argatroban and fondaparinux
in a child with heparin-induced thrombocytopenia. Pediatr
Blood Cancer. 2004;42(suppl):1756.
179. Maurer SH, Wilimas JA, Wang WC, Reiss UM. Heparin
induced thrombocytopenia and re-thrombosis associated
with warfarin and fondaparinux in a child. Pediatr Blood
Cancer. 2009;53(3):468-471.
180. Grabowski E, Bussel J. Pediatric experience with fondaparinux in deep venous thrombosis [ASH Annual Meeting
Abstracts]. Blood. 2006;108:916.
181. Young G. New anticoagulants in children. Hematology
(Am Soc Hematol Educ Program). 2008;208(1):245-250.
182. Rajasekhar D, Barnard MR, Bednarek FJ, Michelson AD.
Platelet hyporeactivity in very low birth weight neonates.
Thromb Haemost. 1997;77(5):1002-1007.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e785S

183. Rajasekhar D, Kestin AS, Bednarek FJ, Ellis PA, Barnard MR,
Michelson AD. Neonatal platelets are less reactive than
adult platelets to physiological agonists in whole blood.
Thromb Haemost. 1994;72(6):957-963.
184. Katz JA , Moake JL , McPherson PD , et al . Relationship
between human development and disappearance of unusually
large von Willebrand factor multimers from plasma. Blood.
1989;73(7):1851-1858.
185. Tsao CH, Green D, Schultz K. Function and ultrastructure
of platelets of neonates: enhanced ristocetin aggregation of
neonatal platelets. Br J Haematol. 1976;32(2):225-233.
186. Weinstein MJ, Blanchard R, Moake JL, Vosburgh E,
Moise K. Fetal and neonatal von Willebrand factor (vWF)
is unusually large and similar to the vWF in patients with
thrombotic thrombocytopenic purpura. Br J Haematol. 1989;
72(1):68-72.
187. Sanders JM, Holtkamp CA, Buchanan GR. The bleeding
time may be longer in children than in adults. Am J Pediatr
Hematol Oncol. 1990;12(3):314-318.
188. Aversa LA, Vzquez A, Pealver JA, Dascal E, Bustelo PM.
Bleeding time in normal children. J Pediatr Hematol Oncol.
1995;17(1):25-28.
189. Carcao MD, Blanchette VS, Dean JA, et al. The Platelet
Function Analyzer (PFA-100): a novel in-vitro system for
evaluation of primary haemostasis in children. Br J Haematol.
1998;101(1):70-73.
190. Israels SJ, Cheang T, McMillan-Ward EM, Cheang M.
Evaluation of primary hemostasis in neonates with a new
in vitro platelet function analyzer. J Pediatr. 2001;138(1):
116-119.
191. Roschitz B, Sudi K, Kstenberger M, Muntean W. Shorter
PFA-100 closure times in neonates than in adults: role of
red cells, white cells, platelets and von Willebrand factor.
Acta Paediatr. 2001;90(6):664-670.
192. Halimeh S, Angelis G, Sander A, et al. Multiplate whole
blood impedance point of care aggregometry: preliminary
reference values in healthy infants, children and adolescents. Klin Padiatr. 2010;222(3):158-163.
193. Israels SJ, Michelson AD. Antiplatelet therapy in children.
Thromb Res. 2006;118(1):75-83.
194. Lietman PS, Done AK, Yaffe SJ, Clayton JM. Aspirin dosage
for infants and children. J Pediatr. 1979;95(4):617-625.
195. Michelson AD. Platelet function testing in cardiovascular
diseases. Circulation. 2004;110(19):e489-e493.
196. Israels SJ, Rand ML, Michelson AD. Neonatal platelet function. Semin Thromb Hemost. 2003;29(4):363-372.
197. Baum J. Aspirin in the treatment of juvenile arthritis. Am
J Med. 1983;74(6A):10-15.
198. Halpin TJ, Holtzhauer FJ, Campbell RJ, et al. Reyes
syndrome and medication use. JAMA. 1982;248(6):687-691.
199. Porter JD, Robinson PH, Glasgow JF, Banks JH, Hall SM.
Trends in the incidence of Reyes syndrome and the use of
aspirin. Arch Dis Child. 1990;65(8):826-829.
200. Remington PL, Shabino CL, McGee H, Preston G,
Sarniak AP, Hall WN. Reye syndrome and juvenile rheumatoid arthritis in Michigan. Am J Dis Child. 1985;139(9):
870-872.
201. Starko KM, Ray CG, Dominguez LB, Stromberg WL,
Woodall DF. Reyes syndrome and salicylate use. Pediatrics.
1980;66(6):859-864.
202. Young RS, Torretti D, Williams RH, Hendriksen D,
Woods M. Reyes syndrome associated with long-term aspirin therapy. JAMA. 1984;251(6):754-756.
203. el Makhlouf A, Friedli B, Oberhnsli I, Rouge JC, Faidutti B.
Prosthetic heart valve replacement in children. Results and
follow-up of 273 patients. J Thorac Cardiovasc Surg. 1987;
93(1):80-85.
e786S

204. LeBlanc JG, Sett SS, Vince DJ. Antiplatelet therapy in children
with left-sided mechanical prostheses. Eur J Cardiothorac
Surg. 1993;7(4):211-215.
205. Solymar L, Rao PS, Mardini MK, Fawzy ME, Guinn G.
Prosthetic valves in children and adolescents. Am Heart J.
1991;121(2 pt 1):557-568.
206. Li JS, Yow E, Berezny KY, et al; PICOLO Investigators.
Dosing of clopidogrel for platelet inhibition in infants and
young children: primary results of the Platelet Inhibition
in Children On cLOpidogrel (PICOLO) trial. Circulation.
2008;117(4):553-559.
207. Mertens L, Eyskens B, Boshoff D, Gewillig M. Safety
and efficacy of clopidogrel in children with heart disease.
J Pediatr. 2008;153(1):61-64.
208. Maltz LA, Gauvreau K, Connor JA, Jenkins KJ. Clopidogrel
in a pediatric population: prescribing practice and outcomes
from a single center. Pediatr Cardiol. 2009;30(2):99-105.
209. Forrester MB. Pattern of clopidogrel exposures reported to
Texas poison centers during 1998-2004. Clin Toxicol (Phila).
2007;45(8):950-955.
210. Coller BS. Blockade of platelet GPIIb/IIIa receptors as an
antithrombotic strategy. Circulation. 1995;92(9):2373-2380.
211. Williams RV, Wilke VM, Tani LY, Minich LL. Does abciximab enhance regression of coronary aneurysms resulting
from Kawasaki disease? Pediatrics. 2002;109(1):E4.
212. Andrew M, Paes B, Johnston M. Development of the hemostatic system in the neonate and young infant. Am J Pediatr
Hematol Oncol. 1990;12(1):95-104.
213. Andrew M, Paes B, Milner R, et al. Development of the
human coagulation system in the full-term infant. Blood.
1987;70(1):165-172.
214. Reverdiau-Moalic P, Gruel Y, Delahousse B, et al. Comparative study of the fibrinolytic system in human fetuses
and in pregnant women. Thromb Res. 1991;61(5-6):489-499.
215. Corrigan JJ Jr, Sleeth JJ, Jeter M, Lox CD. Newborns
fibrinolytic mechanism: components and plasmin generation. Am J Hematol. 1989;32(4):273-278.
216. Leaker M, Massicotte MP, Brooker LA, Andrew M. Thrombolytic therapy in pediatric patients: a comprehensive review
of the literature. Thromb Haemost. 1996;76(2):132-134.
217. Albisetti M. Thrombolytic therapy in children. Thromb Res.
2006;118(1):95-105.
218. Nowak-Gttl U, Junker R, Hartmeier M, et al. Increased
lipoprotein(a) is an important risk factor for venous thromboembolism in childhood. Circulation. 1999;100(7):743-748.
219. Williams MD. Thrombolysis in children. Br J Haematol.
2010;148(1):26-36.
220. Yee DL, Chan AK, Williams S, Goldenberg NA,
Massicotte MP, Raffini LJ. Varied opinions on thrombolysis
for venous thromboembolism in infants and children: findings from a survey of pediatric hematology-oncology specialists. Pediatr Blood Cancer. 2009;53(6):960-966.
221. Public Health Service; Food and Drug Administration.
Important Drug Warning: Safety Information Regarding
the Use of Abbokinase (Urokinase). Rockville, MD: Food
and Drug Administration; 1999.
222. Gupta AA, Leaker M, Andrew M, et al. Safety and outcomes of thrombolysis with tissue plasminogen activator for
treatment of intravascular thrombosis in children. J Pediatr.
2001;139(5):682-688.
223. Raffini L. Thrombolysis for intravascular thrombosis in neonates and children. Curr Opin Pediatr. 2009;21(1):9-14.
224. Newall F, Browne M, Savoia H, Campbell J, Barnes C,
Monagle P. Assessing the outcome of systemic tissue plasminogen activator for the management of venous and
arterial thrombosis in paediatrics. J Pediatr Hematol Oncol.
2007;29(4):269-273.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

225. Goldenberg NA, Durham JD, Knapp-Clevenger R, MancoJohnson MJ. A thrombolytic regimen for high-risk deep
venous thrombosis may substantially reduce the risk of
postthrombotic syndrome in children. Blood. 2007;110(1):
45-53.
226. Manco-Johnson MJ, Grabowski EF, Hellgreen M, et al.
Recommendations for tPA thrombolysis in children. On behalf
of the Scientific Subcommittee on Perinatal and Pediatric
Thrombosis of the Scientific and Standardization Committee
of the International Society of Thrombosis and Haemostasis.
Thromb Haemost. 2002;88(1):157-158.
227. Ade-Ajayi N, Hall NJ, Liesner R, et al. Acute neonatal arterial occlusion: is thrombolysis safe and effective? J Pediatr
Surg. 2008;43(10):1827-1832.
228. Giuffr B, Compagnoni G, Farina C, Mosca F. Successful use
of tissue plasminogen activator (t-PA) in catheter-related
intracardiac thrombi of two premature infants. Acta Paediatr.
1998;87(6):695-698.
229. Hartmann J, Hussein A, Trowitzsch E, Becker J, Hennecke
KH. Treatment of neonatal thrombus formation with recombinant tissue plasminogen activator: six years experience and
review of the literature. Arch Dis Child Fetal Neonatal Ed.
2001;85(1):F18-F22.
230. Watkins S, Yunge M, Jones D, Kiely E, Petros AJ. Prolonged
use of tissue plasminogen activator for bilateral lower limb
arterial occlusion in a neonate. J Pediatr Surg. 2001;36(4):
654-656.
231. Weiner GM, Castle VP, DiPietro MA, Faix RG. Successful
treatment of neonatal arterial thromboses with recombinant
tissue plasminogen activator. J Pediatr. 1998;133(1):133-136.
232. Zenz W, Arlt F, Sodia S, Berghold A. Intracerebral hemorrhage during fibrinolytic therapy in children: a review of
the literature of the last thirty years. Semin Thromb Hemost.
1997;23(3):321-332.
233. Bovill EG, Becker R, Tracy RP. Monitoring thrombolytic
therapy. Prog Cardiovasc Dis. 1992;34(4):279-294.
234. Farnoux C, Camard O, Pinquier D, et al. Recombinant tissue-type plasminogen activator therapy of thrombosis in 16
neonates. J Pediatr. 1998;133(1):137-140.
235. Levy M, Benson LN, Burrows PE, et al. Tissue plasminogen
activator for the treatment of thromboembolism in infants
and children. J Pediatr. 1991;118(3):467-472.
236. Manco-Johnson MJ, Nuss R, Hays T, Krupski W, Drose J,
Manco-Johnson ML. Combined thrombolytic and anticoagulant therapy for venous thrombosis in children. J Pediatr.
2000;136(4):446-453.
237. Wang M, Hays T, Balasa V, et al; Pediatric Coagulation
Consortium. Low-dose tissue plasminogen activator thrombolysis in children. J Pediatr Hematol Oncol. 2003;25(5):
379-386.
238. Zenz W, Muntean W, Beitzke A, Zobel G, Riccabona M,
Gamillscheg A. Tissue plasminogen activator (alteplase)
treatment for femoral artery thrombosis after cardiac catheterisation in infants and children. Br Heart J. 1993;70(4):
382-385.
239. Barzaghi A, DellOrto M, Rovelli A, Rizzari C, Colombini A,
Uderzo C. Central venous catheter clots: incidence, clinical
significance and catheter care in patients with hematologic
malignancies. Pediatr Hematol Oncol. 1995;12(3):243-250.
240. Chesler L, Feusner JH. Use of tissue plasminogen activator
(rt-PA) in young children with cancer and dysfunctional
central venous catheters. J Pediatr Hematol Oncol. 2002;
24(8):653-656.
241. Choi M, Massicotte MP, Marzinotto V, Chan AK, Holmes JL,
Andrew M. The use of alteplase to restore patency of central
venous lines in pediatric patients: a cohort study. J Pediatr.
2001;139(1):152-156.
www.chestpubs.org

242. Curnow A, Idowu J, Behrens E, Toomey F, Georgeson K.


Urokinase therapy for Silastic catheter-induced intravascular thrombi in infants and children. Arch Surg. 1985;
120(11):1237-1240.
243. Haire WD, Deitcher SR, Mullane KM, et al. Recombinant
urokinase for restoration of patency in occluded central
venous access devices. A double-blind, placebo-controlled
trial. Thromb Haemost. 2004;92(3):575-582.
244. Jacobs BR, Haygood M, Hingl J. Recombinant tissue
plasminogen activator in the treatment of central venous
catheter occlusion in children. J Pediatr. 2001;139(4):
593-596.
245. Shen V, Li X, Murdock M, Resnansky L, McCluskey ER,
Semba CP; COOL Investigators. Recombinant tissue plasminogen activator (alteplase) for restoration of function
to occluded central venous catheters in pediatric patients.
J Pediatr Hematol Oncol. 2003;25(1):38-45.
246. Svoboda P, Barton RP, Barbarash OL, et al. Recombinant
urokinase is safe and effective in restoring patency to
occluded central venous access devices: a multiple-center,
international trial. Crit Care Med. 2004;32(10):1990-1996.
247. Wachs T. Urokinase administration in pediatric patients
with occluded central venous catheters. J Intraven Nurs.
1990;13(2):100-102.
248. Wever ML, Liem KD, Geven WB, Tanke RB. Urokinase
therapy in neonates with catheter related central venous
thrombosis. Thromb Haemost. 1995;73(2):180-185.
249. Monagle P, Phelan E, Downie P, et al. Local thrombolytic
therapy in children. Thromb Haemost. 1997;77(suppl):504.
250. Cannizzaro V, Berger F, Kretschmar O, Saurenmann R,
Knirsch W, Albisetti M. Thrombolysis of venous and arterial
thrombosis by catheter-directed low-dose infusion of tissue
plasminogen activator in children. J Pediatr Hematol Oncol.
2005;27(12):688-691.
251. Janmaat M, Gravendeel JP, Uyttenboogaart M, Vroomen PC,
Brouwer OF, Luijckx GJ. Local intra-arterial thrombolysis
in a 4-year-old male with vertebrobasilar artery thrombosis.
Dev Med Child Neurol. 2009;51(2):155-158.
252. Feng X, Jing ZP, Hou JG, Gao X. Prevention of tumor
emboli from the inferior vena cava by the Tempofilter II
during resection of nephroblastoma with level III tumor
thrombus. Chin Med J (Engl). 2010;123(2):253-255.
253. Inoue Y, Kato M, Ohsuka T, Morikawa A. Successful treatment of a child with inferior vena cava thrombosis using a
temporary inferior vena cava filter. Pediatr Cardiol. 2002;
23(1):74-76.
254. Khong PL, John PR. Technical aspects of insertion and
removal of an inferior vena cava IVC filter for prophylactic treatment of pulmonary embolus. Pediatr Radiol.
1997;27(3):239-241.
255. McBride WJ, Gadowski GR, Keller MS, Vane DW. Pulmonary embolism in pediatric trauma patients. J Trauma.
1994;37(6):913-915.
256. Reed RA, Teitelbaum GP, Stanley P, Mazer MJ, Tonkin IL,
Rollins NK. The use of inferior vena cava filters in pediatric patients for pulmonary embolus prophylaxis. Cardiovasc
Intervent Radiol. 1996;19(6):401-405.
257. Williams S, Chait P, Temple M. Vena cava filters in children:
review of a single centre clinical experience over 17 years.
Thromb Haemost. 2003;1):OC439.
258. Mody RN, Stokes LS, Bream PR Jr, Spottswood SE.
Removal of a Gnther Tulip retrievable inferior vena cava
filter after 147 days in a pediatric patient. Pediatr Radiol.
2006;36(5):440-444.
259. Raffini L, Cahill AM, Hellinger J, Manno C. A prospective observational study of IVC filters in pediatric patients.
Pediatr Blood Cancer. 2008;51(4):517-520.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e787S

260. Chaudry G, Padua HM, Alomari AI. The use of inferior


vena cava filters in young children. J Vasc Interv Radiol.
2008;19(7):1103-1106.
261. Arshad A, McCarthy MJ. Management of limb ischaemia
in the neonate and infant. Eur J Vasc Endovasc Surg. 2009;
38(1):61-65.
262. Choi SH, Jeong SI, Yang JH, et al. A single-center experience with intracardiac thrombosis in children with dilated
cardiomyopathy. Pediatr Cardiol. 2010;31(2):264-269.
263. Coombs CJ, Richardson PW, Dowling GJ, Johnstone BR,
Monagle P. Brachial artery thrombosis in infants: an algorithm
for limb salvage. Plast Reconstr Surg. 2006;117(5):1481-1488.
264. Gossett JG, Rocchini AP, Armstrong AK. Superior vena
cava thrombectomy with the X-SIZER catheter system in
a child with Fontan palliation. Catheter Cardiovasc Interv.
2007;69(1):28-32.
265. Gu C, Fan S, Zhou H, et al. Surgical treatment of giant coronary artery aneurysm secondary to Kawasaki disease. Heart
Surg Forum. 2009;12(4):E241-E243.
266. Kokov LS, Korostelev AN, Grinko AN, et al. Recanalization
and thrombectomy of internal anastomosis in a patient with
tetralogy of Fallot using the AngioJet rheolytic catheter.
Catheter Cardiovasc Interv. 2001;53(4):504-507.
267. Lodge AJ, Jaggers J, Adams D, Rice HE. Vascular control
for resection of suprahepatic intracaval Wilms tumor: technical considerations. J Pediatr Surg. 2000;35(12):1836-1837.
268. Martnez-Ibez V, Snchez de Toledo J, De Diego M, et al.
Wilms tumours with intracaval involvement. Med Pediatr
Oncol. 1996;26(4):268-271.
269. Menon SC, Hagler DJ, Cetta F, Cabalka AK. Rheolytic
mechanical thrombectomy for pulmonary artery thrombus
in children with complex cyanotic congenital heart disease.
Catheter Cardiovasc Interv. 2008;71(2):237-243.
270. Ries M, Zenker M, Girisch M, Klinge J, Singer H. Percutaneous endovascular catheter aspiration thrombectomy
of severe superior vena cava syndrome. Arch Dis Child Fetal
Neonatal Ed. 2002;87(1):F64-F66.
271. Sarigl A, Farsak B, Koramaz I, Tok M, Yurdakul Y.
Postoperative graft thrombosis in Fontan procedure. Turk
J Pediatr. 2000;42(1):80-83.
272. Sur JP, Garg RK, Jolly N. Rheolytic percutaneous thrombectomy for acute pulmonary embolism in a pediatric patient.
Catheter Cardiovasc Interv. 2007;70(3):450-453.
273. Yang JY, Williams S, Brando LR, Chan AK. Neonatal and
childhood right atrial thrombosis: recognition and a riskstratified treatment approach. Blood Coagul Fibrinolysis.
2010;21(4):301-307.
274. Obladen M, Ernst D, Feist D, Wille L. Portal hypertension
in children following neonatal umbilical disorders. J Perinat
Med. 1975;3(2):101-104.
275. Vos LJ, Potocky V, Brker FH, de Vries JA, Postma L, Edens
E. Splenic vein thrombosis with oesophageal varices: a late
complication of umbilical vein catheterization. Ann Surg.
1974;180(2):152-156.
276. Filippi L, Palermo L, Pezzati M, et al. Paradoxical embolism
in a preterm infant. Dev Med Child Neurol. 2004;46(10):
713-716.
277. Bkenkamp A, von Kries R, Nowak-Gttl U, Gbel U,
Hoyer PF. Neonatal renal venous thrombosis in Germany
between 1992 and 1994: epidemiology, treatment and outcome. Eur J Pediatr. 2000;159(1-2):44-48.
278. Zigman A, Yazbeck S, Emil S, Nguyen L. Renal vein thrombosis: a 10-year review. J Pediatr Surg. 2000;35(11):1540-1542.
279. Lau KK, Stoffman JM, Williams S, et al; Canadian Pediatric Thrombosis and Hemostasis Network. Neonatal renal
vein thrombosis: review of the English-language literature
between 1992 and 2006. Pediatrics. 2007;120(5):e1278-e1284.
e788S

280. Mocan H, Beattie TJ, Murphy AV. Renal venous thrombosis


in infancy: long-term follow-up. Pediatr Nephrol. 1991;5(1):
45-49.
281. Nuss R, Hays T, Manco-Johnson M. Efficacy and safety of
heparin anticoagulation for neonatal renal vein thrombosis.
Am J Pediatr Hematol Oncol. 1994;16(2):127-131.
282. Ricci MA, Lloyd DA. Renal venous thrombosis in infants
and children. Arch Surg. 1990;125(9):1195-1199.
283. Kosch A, Kuwertz-Brking E, Heller C, Kurnik K,
Schobess R, Nowak-Gttl U. Renal venous thrombosis in
neonates: prothrombotic risk factors and long-term follow-up.
Blood. 2004;104(5):1356-1360.
284. Shah PS, Shah VS. Continuous heparin infusion to prevent thrombosis and catheter occlusion in neonates with
peripherally placed percutaneous central venous catheters.
Cochrane Database Syst Rev. 2008;(2):CD002772.
285. Barrington KJ. Umbilical artery catheters in the newborn:
effects of position of the catheter tip. Cochrane Database
Syst Rev. 2000;(2):CD000505.
286. Kamala F, Boo NY, Cheah FC, Birinder K. Randomized
controlled trial of heparin for prevention of blockage of
peripherally inserted central catheters in neonates. Acta
Paediatr. 2002;91(12):1350-1356.
287. Birch P, Ogden S, Hewson M. A randomised, controlled
trial of heparin in total parenteral nutrition to prevent sepsis associated with neonatal long lines: the Heparin in Long
Line Total Parenteral Nutrition (HILLTOP) trial. Arch Dis
Child Fetal Neonatal Ed. 2010;95(4):F252-F257.
288. Tsai KT, Chang CH, Lin PJ. Modified Blalock-Taussig shunt:
statistical analysis of potential factors influencing shunt outcome. J Cardiovasc Surg (Torino). 1996;37(2):149-152.
289. Gedicke M, Morgan G, Parry A, Martin R, Tulloh R. Risk
factors for acute shunt blockage in children after modified Blalock-Taussig shunt operations. Heart Vessels. 2010;
25(5):405-409.
290. Ahmad U, Fatimi SH, Naqvi I, et al. Modified Blalock-Taussig
shunt: immediate and short-term follow-up results in neonates. Heart Lung Circ. 2008;17(1):54-58.
291. Wells WJ, Yu RJ, Batra AS, Monforte H, Sintek C,
Starnes VA. Obstruction in modified Blalock shunts: a
quantitative analysis with clinical correlation. Ann Thorac
Surg. 2005;79(6):2072-2076.
292. Fenton KN, Siewers RD, Rebovich B, Pigula FA. Interim
mortality in infants with systemic-to-pulmonary artery shunts.
Ann Thorac Surg. 2003;76(1):152-156, discussion 156-157.
293. Al Jubair KA, Al Fagih MR, Al Jarallah AS, et al. Results
of 546 Blalock-Taussig shunts performed in 478 patients.
Cardiol Young. 1998;8(4):486-490.
294. Li JS, Yow E, Berezny KY, et al. Clinical outcomes of palliative surgery including a systemic-to-pulmonary artery
shunt in infants with cyanotic congenital heart disease:
does aspirin make a difference? Circulation. 2007;116(3):
293-297.
295. Motz R, Wessel A, Ruschewski W, Brsch J. Reduced
frequency of occlusion of aorto-pulmonary shunts in infants
receiving aspirin. Cardiol Young. 1999;9(5):474-477.
296. Mullen JC, Lemermeyer G, Bentley MJ. Modified BlalockTaussig shunts: to heparinize or not to heparinize? Can J
Cardiol. 1996;12(7):645-647.
297. MacMillan M, Jones TK, Lupinetti FM, Johnston TA.
Balloon angioplasty for Blalock-Taussig shunt failure in the
early postoperative period. Catheter Cardiovasc Interv.
2005;66(4):585-589.
298. Sreeram N, Emmel M, Ben-Mime L, Brockmeier K,
Bennink G. Transcatheter recanalization of acutely occluded
modified systemic to pulmonary artery shunts in infancy.
Clin Res Cardiol. 2008;97(3):181-186.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

299. Wang JK, Wu MH, Chang CI, Chiu IS, Lue HC. Balloon
angioplasty for obstructed modified systemic-pulmonary
artery shunts and pulmonary artery stenoses. J Am Coll
Cardiol. 2001;37(3):940-947.
300. Chang YM, Huang SC, Chen SJ, et al. An unusual cause of
chylothorax after Norwood stage one reconstruction. Thorac
Cardiovasc Surg. 2009;57(2):120-122.
301. Duncan WJ, Campbell AI, Human DG. Left ventricular
thrombosis following a Norwood procedure. Cardiol Young.
2007;17(2):232.
302. Sano S, Huang SC, Kasahara S, Yoshizumi K, Kotani Y,
Ishino K. Risk factors for mortality after the Norwood procedure using right ventricle to pulmonary artery shunt. Ann
Thorac Surg. 2009;87(1):178-186..
303. Celermajer DS, Robinson JT, Taylor JF. Vascular access
in previously catheterised children and adolescents: a prospective study of 131 consecutive cases. Br Heart J. 1993;
70(6):554-557.
304. Hurwitz RA, Franken EA Jr, Girod DA, Smith JA, Smith WL.
Angiographic determination of arterial patency after percutaneous catheterization in infants and small children.
Circulation. 1977;56(1):102-105.
305. Kern IB. Management of children with chronic femoral
artery obstruction. J Pediatr Surg. 1977;12(1):83-90.
306. Taylor LM Jr, Troutman R, Feliciano P, Menashe V,
Sunderland C, Porter JM. Late complications after femoral
artery catheterization in children less than five years of age.
J Vasc Surg. 1990;11(2):297-304.
307. Brus F, Witsenburg M, Hofhuis WJ, Hazelzet JA, Hess J.
Streptokinase treatment for femoral artery thrombosis after
arterial cardiac catheterisation in infants and children. Br
Heart J. 1990;63(5):291-294.
308. Carlson KM, Rutledge JM, Parker BR, Grifka RG. Use of
tissue plasminogen activator for femoral artery thrombosis
following transcatheter coil occlusion of patent ductus arteriosus. Pediatr Cardiol. 2005;26(1):83-86.
309. Friedman J, Fabre J, Netscher D, Jaksic T. Treatment
of acute neonatal vascular injuriesthe utility of multiple
interventions. J Pediatr Surg. 1999;34(6):940-945.
310. Giacoia GP. High-dose urokinase therapy in newborn infants
with major vessel thrombosis. Clin Pediatr (Phila). 1993;
32(4):231-237.
311. Grich J, Rilinger N, Sokiranski R, et al. Mechanical thrombolysis of acute occlusion of both the superficial and the
deep femoral arteries using a thrombectomy device. AJR
Am J Roentgenol. 1998;170(5):1177-1180.
312. Harrison BM, Wood CB. Spontaneous femoral artery thrombosis and intermittent claudication in childhood nephrotic
syndrome. Arch Dis Child. 1972;47(255):836-837.
313. Ino T, Benson LN, Freedom RM, Barker GA, Aipursky A,
Rowe RD. Thrombolytic therapy for femoral artery thrombosis after pediatric cardiac catheterization. Am Heart J.
1988;115(3):633-639.
314. Kobayashi T, Kobayashi T, Shinohara M, Tomomasa T,
Morikawa A. Percutaneous hydrodynamic thrombectomy
for femoral arterial thrombosis after arterial catheterization.
Pediatr Cardiol. 2003;24(4):409-411.
315. Kothari SS, Kumar RK, Varma S, Saxena A. Thrombolytic
therapy in infants for femoral artery thrombosis following cardiac catheterisation. Indian Heart J. 1996;48(3):
246-248.
316. Lin PH, Dodson TF, Bush RL, et al. Surgical intervention
for complications caused by femoral artery catheterization
in pediatric patients. J Vasc Surg. 2001;34(6):1071-1078.
317. Lincoln JC, Deverall PB. The treatment of arterial thrombosis in infants and children by balloon catheters. J Pediatr
Surg. 1969;4(3):359-362.
www.chestpubs.org

318. Liu Q, Yan CW, Zhao SH, et al. Thrombolytic therapy for
femoral artery thrombosis after left cardiac catheterization
in children. Chin Med J (Engl). 2009;122(8):931-934.
319. Mack RM, Hartmann JR, Sauvage LR. Iliofemoral venous
thrombectomy in a child with a coagulation abnormality.
J Pediatr. 1966;68(3):374-380.
320. Ries M, Singer H, Hofbeck M, Klinge J. Tissue plasminogen
activator (alteplase) treatment for femoral artery thrombosis after cardiac catheterisation in infants and children. Br
Heart J. 1994;72(4):403.
321. Wessel DL, Keane JF, Fellows KE, Robichaud H, Lock JE.
Fibrinolytic therapy for femoral arterial thrombosis after
cardiac catheterization in infants and children. Am J Cardiol.
1986;58(3):347-351.
322. Salvino MJ, Ramaswamy R, Schechter LS. Microvascular
reconstruction of iatrogenic femoral artery thrombosis in
an infant: a case report and review of the literature: infant
femoral artery reconstruction. Eplasty. 2009;9:e20.
323. Butt W, Shann F, McDonnell G, Hudson I. Effect of heparin concentration and infusion rate on the patency of arterial catheters. Crit Care Med. 1987;15(3):230-232.
324. Rais-Bahrami K, Karna P, Dolanski EA. Effect of fluids on
life span of peripheral arterial lines. Am J Perinatol. 1990;
7(2):122-124.
325. Selldn H, Nilsson K, Larsson LE, Ekstrm-Jodal B. Radial
arterial catheters in children and neonates: a prospective
study. Crit Care Med. 1987;15(12):1106-1109.
326. Heulitt MJ, Farrington EA, OShea TM, Stoltzman SM,
Srubar NB, Levin DL. Double-blind, randomized, controlled trial of papaverine-containing infusions to prevent
failure of arterial catheters in pediatric patients. Crit Care
Med. 1993;21(6):825-829.
327. Aslam M, Guglietti D, Hansen AR. Neonatal arterial
thrombosis at birth: case report and literature review. Am J
Perinatol. 2008;25(6):347-352.
328. Tarry WC, Moser AJ, Makhoul RG. Peripheral arterial
thrombosis in the nephrotic syndrome. Surgery. 1993;
114(3):618-623.
329. Albisetti M, Schmugge M, Haas R, et al. Arterial thromboembolic complications in critically ill children. J Crit Care.
2005;20(3):296-300.
330. Alpert J, ODonnell JA, Parsonnet V, Brief DK, Brener BJ,
Goldenkranz RJ. Clinically recognized limb ischemia in the
neonate after umbilical artery catheterization. Am J Surg.
1980;140(3):413-418.
331. ONeill JA Jr, Neblett WW III, Born ML. Management of
major thromboembolic complications of umbilical artery
catheters. J Pediatr Surg. 1981;16(6):972-978.
332. Stringel G, Mercer S, Richler M, McMurray B. Catheterization of the umbilical artery in neonates: surgical implications. Can J Surg. 1985;28(2):143-146.
333. Boo NY, Wong NC, Zulkifli SS, Lye MS. Risk factors associated with umbilical vascular catheter-associated thrombosis in
newborn infants. J Paediatr Child Health. 1999;35(5):460-465.
334. Joseph R, Chong A, Teh M, Wee A, Tan KL. Thrombotic
complication of umbilical arterial catheterization and its
sequelae. Ann Acad Med Singapore. 1985;14(4):576-582.
335. Klinger G, Hellmann J, Daneman A. Severe aortic thrombosis
in the neonatesuccessful treatment with low-molecularweight heparin: two case reports and review of the literature.
Am J Perinatol. 2000;17(3):151-158.
336. Krueger TC, Neblett WW, ONeill JA, MacDonell RC,
Dean RH, Thieme GA. Management of aortic thrombosis
secondary to umbilical artery catheters in neonates. J Pediatr
Surg. 1985;20(4):328-332.
337. Joshi VV, Draper DA, Bates RD III. Neonatal necrotizing
enterocolitis. Occurrence secondary to thrombosis of
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e789S

338.
339.
340.

341.
342.
343.

344.
345.

346.

347.
348.
349.

350.
351.

352.
353.
354.

355.
356.

abdominal aorta following umbilical arterial catheterization. Arch Pathol. 1975;99(10):540-543.


Rand T, Weninger M, Kohlhauser C, et al. Effects of umbilical arterial catheterization on mesenteric hemodynamics.
Pediatr Radiol. 1996;26(7):435-438.
Vailas GN, Brouillette RT, Scott JP, Shkolnik A, Conway J,
Wiringa K. Neonatal aortic thrombosis: recent experience.
J Pediatr. 1986;109(1):101-108.
Seibert JJ, Northington FJ, Miers JF, Taylor BJ. Aortic
thrombosis after umbilical artery catheterization in neonates: prevalence of complications on long-term follow-up.
AJR Am J Roentgenol. 1991;156(3):567-569.
Ankola PA, Atakent YS. Effect of adding heparin in very low
concentration to the infusate to prolong the patency of umbilical artery catheters. Am J Perinatol. 1993;10(3):229-232.
Bosque E, Weaver L. Continuous versus intermittent heparin infusion of umbilical artery catheters in the newborn
infant. J Pediatr. 1986;108(1):141-143.
Chang GY, Lueder FL, DiMichele DM, Radkowski MA,
McWilliams LJ, Jansen RD. Heparin and the risk of intraventricular hemorrhage in premature infants. J Pediatr.
1997;131(3):362-366.
David RJ, Merten DF, Anderson JC, Gross S. Prevention
of umbilical artery catheter clots with heparinized infusates.
Dev Pharmacol Ther. 1981;2(2):117-126.
Horgan MJ, Bartoletti A, Polansky S, Peters JC, Manning
TJ, Lamont BM. Effect of heparin infusates in umbilical
arterial catheters on frequency of thrombotic complications.
J Pediatr. 1987;111(5):774-778.
Rajani K, Goetzman BW, Wennberg RP, Turner E,
Abildgaard C. Effect of heparinization of fluids infused
through an umbilical artery catheter on catheter patency and
frequency of complications. Pediatrics. 1979;63(4):552-556.
Barrington KJ. Umbilical artery catheters in the newborn:
effects of heparin. Cochrane Database Syst Rev. 2000;
(2):CD000507.
McDonald M, Johnson M, Rumack C, et al. Heparin prevention of catheter related thrombosis. Pediatr Res. 1984;
18(suppl):18.
Lesko SM, Mitchell AA, Epstein MF, Louik C, Giacoia GP,
Shapiro S. Heparin use as a risk factor for intraventricular
hemorrhage in low-birth-weight infants. N Engl J Med.
1986;314(18):1156-1160.
Malloy MH, Cutter GR. The association of heparin exposure with intraventricular hemorrhage among very low birth
weight infants. J Perinatol. 1995;15(3):185-191.
Bjarke B, Herin P, Blombck M. Neonatal aortic thrombosis. A possible clinical manifestation of congenital antithrombin 3 deficiency. Acta Paediatr Scand. 1974;63(2):
297-301.
Corrigan JJ Jr, Jeter M, Allen HD, Malone JM. Aortic
thrombosis in a neonate: failure of urokinase thrombolytic
therapy. Am J Pediatr Hematol Oncol. 1982;4(3):243-247.
Francis JV, Monagle P, Hope S, Sehgal A. Occlusive aortic arch thrombus in a preterm neonate. Pediatr Crit Care
Med. 2010;11(1):e13-e15.
Janssen DR, Ohmstede DP, Liske MR, Parra D, Drinkwater
D, Kavanaugh-McHugh A. Thromboses in the native aorta
in patients with hypoplastic left heart syndrome. Congenit
Heart Dis. 2007;2(1):74-78.
Lanari M, Lazzarotto T, Papa I, et al. Neonatal aortic arch
thrombosis as a result of congenital cytomegalovirus infection. Pediatrics. 2001;108(6):E114.
Newman RS, Spear GS, Kirschbaum N. Postmortem DNA
diagnosis of factor V Leiden in a neonate with systemic
thrombosis and probable antithrombin deficiency. Obstet
Gynecol. 1998;92(4 pt 2):702-705.

e790S

357. Nouri S, Mahdhaoui N, Beizig S, et al. Major neonatal


aortic thrombosis: a case report [in French]. Arch Pediatr.
2007;14(9):1097-1100.
358. Snchez J, Velasco F, Alvarez R, Romn J, Torres A. Aortic
thrombosis in a neonate with hereditary antithrombin III
deficiency: successful outcome with thrombolytic and
replacement treatment. Acta Paediatr. 1996;85(2):245-247.
359. Sheridan-Pereira M, Porreco RP, Hays T, Burke MS.
Neonatal aortic thrombosis associated with the lupus anticoagulant. Obstet Gynecol. 1988;71(6 pt 2):1016-1018.
360. Tugrul Kural I, Tinaztepe K, Yurdakul Y. Aortic thrombosis in the newborn infant. J Cardiovasc Surg (Torino).
1984;25(3):246-248.
361. Tuohy J, Harrison A. Prenatal transfer of anticardiolipin
antibodies associated with fatal neonatal aortic thrombosis.
Aust N Z J Obstet Gynaecol. 2005;45(2):175-176.
362. Hamilton RM, Penkoske PA, Byrne P, Duncan NF.
Spontaneous aortic thrombosis in a neonate presenting as
coarctation. Ann Thorac Surg. 1988;45(5):564-565.
363. Nagel K, Tuckuviene R, Paes B, Chan AK. Neonatal aortic thrombosis: a comprehensive review. Klin Padiatr.
2010;222(3):134-139.
364. Ahluwalia JS, Kelsall AW, Diederich S, Rennie JM. Successful treatment of aortic thrombosis after umbilical catheterization with tissue plasminogen activator. Acta Paediatr.
1994;83(11):1215-1217.
365. Colburn MD, Gelabert HA, Quiones-Baldrich W. Neonatal
aortic thrombosis. Surgery. 1992;111(1):21-28.
366. Kawahira Y, Kishimoto H, Lio M, et al. Spontaneous aortic thrombosis in a neonate with multiple thrombi in the
main branches of the abdominal aorta. Cardiovasc Surg.
1995;3(2):219-221.
367. Kothari SS, Varma S, Wasir HS. Thrombolytic therapy in
infants and children. Am Heart J. 1994;127(3):651-657.
368. Sahoo S, Das PK. A neonate with complete thrombosis of
the aorta. Indian J Pediatr. 2009;76(5):563-564.
369. Sharathkumar AA, Lamear N, Pipe S, et al. Management
of neonatal aortic arch thrombosis with low-molecular
weight heparin: a case series. J Pediatr Hematol Oncol.
2009;31(7):516-521.
370. Sherman GG, Mnster M, Govendrageloo K, Harrisberg J,
Levin SE. Low molecular weight heparin in the successful
treatment of a spontaneous aortic thrombosis in a neonate.
Pediatr Hematol Oncol. 2000;17(5):409-413.
371. Torkington J, Hitchcock R, Wilkinson K, Kiely E. Successful
use of recombinant tissue plasminogen activator in the treatment of aortic thrombosis in a premature neonate. Eur J
Vasc Endovasc Surg. 1997;13(5):515-516.
372. Irving C, Zaman A, Kirk R. Transradial coronary angiography in children and adolescents. Pediatr Cardiol.
2009;30(8):1089-1093.
373. Freed MD, Keane JF, Rosenthal A. The use of heparinization to prevent arterial thrombosis after percutaneous cardiac
catheterization in children. Circulation. 1974;50(3):565-569.
374. Freed MD, Rosenthal A, Fyler D. Attempts to reduce
arterial thrombosis after cardiac catheterization in children:
use of percutaneous technique and aspirin. Am Heart J.
1974;87(3):283-286.
375. Girod DA, Hurwitz RA, Caldwell RL. Heparinization for
prevention of thrombosis following pediatric percutaneous
arterial catheterization. Pediatr Cardiol. 1982;3(2):175-180.
376. Rao PS, Thapar MK, Rogers JH Jr, et al. Effect of intraarterial injection of heparin on the complications of percutaneous arterial catheterization in infants and children. Cathet
Cardiovasc Diagn. 1981;7(3):235-246.
377. Saxena A, Gupta R, Kumar RK, Kothari SS, Wasir HS.
Predictors of arterial thrombosis after diagnostic cardiac
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

378.
379.

380.

381.
382.

383.
384.
385.
386.
387.
388.

389.
390.

391.
392.

393.
394.

395.

catheterization in infants and children randomized to two


heparin dosages. Cathet Cardiovasc Diagn. 1997;41(4):
400-403.
Heller C, Heinecke A, Junker R, et al; Childhood Stroke
Study Group. Cerebral venous thrombosis in children: a
multifactorial origin. Circulation. 2003;108(11):1362-1367.
Medlock MD, Olivero WC, Hanigan WC, Wright RM,
Winek SJ. Children with cerebral venous thrombosis diagnosed with magnetic resonance imaging and magnetic resonance angiography. Neurosurgery. 1992;31(5):870-876,
discussion 876.
Laurichesse Delmas H, Winer N, Gallot D, et al. Prenatal
diagnosis of thrombosis of the dural sinuses: report of six
cases, review of the literature and suggested management.
Ultrasound Obstet Gynecol. 2008;32(2):188-198.
Barron TF, Gusnard DA, Zimmerman RA, Clancy RR.
Cerebral venous thrombosis in neonates and children.
Pediatr Neurol. 1992;8(2):112-116.
Wu SL, Amato H, Biringer R, Choudhary G, Shieh P,
Hancock WS. Targeted proteomics of low-level proteins in
human plasma by LC/MSn: using human growth hormone
as a model system. J Proteome Res. 2002;1(5):459-465.
deVeber G, Andrew M, Adams C, et al; Canadian Pediatric
Ischemic Stroke Study Group. Cerebral sinovenous thrombosis in children. N Engl J Med. 2001;345(6):417-423.
Fitzgerald KC, Williams LS, Garg BP, Carvalho KS, Golomb
MR. Cerebral sinovenous thrombosis in the neonate. Arch
Neurol. 2006;63(3):405-409.
Wu YW, Hamrick SE, Miller SP, et al. Intraventricular hemorrhage in term neonates caused by sinovenous thrombosis.
Ann Neurol. 2003;54(1):123-126.
Berfelo FJ, Kersbergen KJ, van Ommen CH, et al. Neonatal
cerebral sinovenous thrombosis from symptom to outcome.
Stroke. 2010;41(7):1382-1388.
Carvalho KS, Bodensteiner JB, Connolly PJ, Garg BP.
Cerebral venous thrombosis in children. J Child Neurol.
2001;16(8):574-580.
deVeber GA, MacGregor D, Curtis R, Mayank S. Neurologic outcome in survivors of childhood arterial ischemic
stroke and sinovenous thrombosis. J Child Neurol. 2000;15(5):
316-324.
Shevell MI, Silver K, OGorman AM, Watters GV, Montes
JL. Neonatal dural sinus thrombosis. Pediatr Neurol.
1989;5(3):161-165.
Moharir M, Shroff M, MacGregor D, et al. Clinical and radiographic features of thrombosis propagation in neonatal
and childhood cerebral sinovenous thrombosis. Ann Neurol.
2006;60(suppl 10):S141.
Ramenghi LA, Govaert P, Fumagalli M, Bassi L, Mosca F.
Neonatal cerebral sinovenous thrombosis. Semin Fetal
Neonatal Med. 2009;14(5):278-283.
Yang JY, Chan AK, Callen DJ, Paes BA. Neonatal cerebral
sinovenous thrombosis: sifting the evidence for a diagnostic plan and treatment strategy. Pediatrics. 2010;126(3):
e693-e700.
Moharir MD, Shroff M, Stephens D, et al. Anticoagulants in
pediatric cerebral sinovenous thrombosis: a safety and outcome study. Ann Neurol. 2010;67(5):590-599.
Jordan LC, Rafay MF, Smith SE, et al. Antithrombotic treatment in neonatal cerebral sinovenous thrombosis: results of
the International Pediatric Stroke Study. J Pediatr. 2010;
156(5):704-710.
Kersbergen KJ, de Vries LS, van Straaten HL, Benders MJ,
Nievelstein RA, Groenendaal F. Anticoagulation therapy
and imaging in neonates with a unilateral thalamic hemorrhage due to cerebral sinovenous thrombosis. Stroke.
2009;40(8):2754-2760.

www.chestpubs.org

396. Kirton A, deVeber G. Advances in perinatal ischemic stroke.


Pediatr Neurol. 2009;40(3):205-214.
397. Dudink J, Mercuri E, Al-Nakib L, et al. Evolution of unilateral perinatal arterial ischemic stroke on conventional and
diffusion-weighted MR imaging. AJNR Am J Neuroradiol.
2009;30(5):998-1004.
398. Tonse R, Nelson KB, Ferriero D, Lynch JK; NICHDNINDS Perinatal Stroke Workshop Participants.
Ischemic perinatal stroke: summary of a workshop sponsored by the National Institute of Child Health and
Development and the National Institute of Neurological
Disorders and. Pediatrics. 2007;120(3):609-616.
399. Lynch JK, Hirtz DG, DeVeber G, Nelson KB. Report of
the National Institute of Neurological Disorders and Stroke
workshop on perinatal and childhood stroke. Pediatrics.
2002;109(1):116-123.
400. Golomb MR, MacGregor DL, Domi T, et al. Presumed
pre- or perinatal arterial ischemic stroke: risk factors and
outcomes. Ann Neurol. 2001;50(2):163-168.
401. Kirton A, Shroff M, Pontigon AM, deVeber G. Risk factors
and presentations of periventricular venous infarction vs
arterial presumed perinatal ischemic stroke. Arch Neurol.
2010;67(7):842-848.
402. Benders MJ, Groenendaal F, Uiterwaal CS, de Vries LS.
Perinatal arterial stroke in the preterm infant. Semin
Perinatol. 2008;32(5):344-349.
403. Cheong JL, Cowan FM. Neonatal arterial ischaemic stroke:
obstetric issues. Semin Fetal Neonatal Med. 2009;14(5):
267-271.
404. Gnther G, Junker R, Strter R, et al; Childhood Stroke
Study Group. Symptomatic ischemic stroke in full-term
neonates: role of acquired and genetic prothrombotic risk
factors. Stroke. 2000;31(10):2437-2441.
405. Lee J, Croen LA, Backstrand KH, et al. Maternal and infant
characteristics associated with perinatal arterial stroke in the
infant. JAMA. 2005;293(6):723-729.
406. Mercuri E, Cowan F, Gupte G, et al. Prothrombotic disorders and abnormal neurodevelopmental outcome in infants
with neonatal cerebral infarction. Pediatrics. 2001;107(6):
1400-1404.
407. Barkat-Masih M, Saha C, Hamby DK, Ofner S, Golomb
MR. Feeding problems in children with neonatal arterial
ischemic stroke. J Child Neurol. 2010;25(7):867-872.
408. Golomb MR. Outcomes of perinatal arterial ischemic stroke
and cerebral sinovenous thrombosis. Semin Fetal Neonatal
Med. 2009;14(5):318-322.
409. Golomb MR, Garg BP, Edwards-Brown M, Williams LS.
Very early arterial ischemic stroke in premature infants.
Pediatr Neurol. 2008;38(5):329-334.
410. Westmacott R, MacGregor D, Askalan R, deVeber G. Late
emergence of cognitive deficits after unilateral neonatal
stroke. Stroke. 2009;40(6):2012-2019.
411. De Vries LS, Van der Grond J, Van Haastert IC,
Groenendaal F. Prediction of outcome in new-born infants
with arterial ischaemic stroke using diffusion-weighted
magnetic resonance imaging. Neuropediatrics. 2005;36(1):
12-20.
412. Husson B, Hertz-Pannier L, Renaud C, et al; AVCnn Group.
Motor outcomes after neonatal arterial ischemic stroke
related to early MRI data in a prospective study. Pediatrics.
2010;126(4):912-918.
413. Fullerton HJ, Wu YW, Sidney S, Johnston SC. Risk of recurrent childhood arterial ischemic stroke in a populationbased cohort: the importance of cerebrovascular imaging.
Pediatrics. 2007;119(3):495-501.
414. Kurnik K, Kosch A, Strter R, Schobess R, Heller C,
Nowak-Gttl U; Childhood Stroke Study Group. Recurrent
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e791S

415.
416.
417.
418.
419.
420.
421.

422.
423.

424.
425.

426.

427.
428.

429.

430.
431.

432.
433.

thromboembolism in infants and children suffering from


symptomatic neonatal arterial stroke: a prospective follow-up
study. Stroke. 2003;34(12):2887-2892.
Salih MA, Abdel-Gader AG, Al-Jarallah AA, Kentab AY,
Al-Nasser MN. Outcome of stroke in Saudi children. Saudi
Med J. 2006;27(suppl 1):S91-S96.
Adcock DM, Brozna J, Marlar RA. Proposed classification
and pathologic mechanisms of purpura fulminans and skin
necrosis. Semin Thromb Hemost. 1990;16(4):333-340.
Adcock DM, Hicks MJ. Dermatopathology of skin necrosis
associated with purpura fulminans. Semin Thromb Hemost.
1990;16(4):283-292.
Auletta MJ, Headington JT. Purpura fulminans. A cutaneous manifestation of severe protein C deficiency. Arch
Dermatol. 1988;124(9):1387-1391.
Goldenberg NA, Manco-Johnson MJ. Protein C deficiency.
Haemophilia. 2008;14(6):1214-1221.
Auberger K. Evaluation of a new protein-C concentrate
and comparison of protein-C assays in a child with congenital
protein-C deficiency. Ann Hematol. 1992;64(3):146-151.
Ben-Tal O, Zivelin A, Seligsohn U. The relative frequency
of hereditary thrombotic disorders among 107 patients
with thrombophilia in Israel. Thromb Haemost. 1989;61(1):
50-54.
Burrows RF, Kelton JG. Low fetal risks in pregnancies
associated with idiopathic thrombocytopenic purpura. Am J
Obstet Gynecol. 1990;163(4 pt 1):1147-1150.
Deguchi K, Tsukada T, Iwasaki E, et al. Late-onset homozygous protein C deficiency manifesting cerebral infarction
as the first symptom at age 27. Intern Med. 1992;31(7):
922-925.
Grundy CB, Melissari E, Lindo V, Scully MF, Kakkar VV,
Cooper DN. Late-onset homozygous protein C deficiency.
Lancet. 1991;338(8766):575-576.
Marlar RA, Adcock DM, Madden RM. Hereditary dysfunctional protein C molecules (type II): assay characterization and proposed classification. Thromb Haemost.
1990;63(3):375-379.
Marlar RA, Montgomery RR, Broekmans AW; Report of
the Working Party on Homozygous Protein C Deficiency
of the ICTH-Subcommittee on Protein C and Protein S.
Report on the diagnosis and treatment of homozygous
protein C deficiency. Thromb Haemost. 1989;61(3):529-531.
Marlar RA, Neumann A. Neonatal purpura fulminans due
to homozygous protein C or protein S deficiencies. Semin
Thromb Hemost. 1990;16(4):299-309.
Marlar RA, Sills RH, Groncy PK, Montgomery RR,
Madden RM. Protein C survival during replacement therapy in homozygous protein C deficiency. Am J Hematol. 1992;
41(1):24-31.
Pescatore P, Horellou HM, Conard J, et al. Problems of
oral anticoagulation in an adult with homozygous protein C
deficiency and late onset of thrombosis. Thromb Haemost.
1993;69(4):311-315.
Petrini P, Segnestam K, Ekelund H, Egberg N. Homozygous
protein C deficiency in two siblings. Pediatr Hematol Oncol.
1990;7(2):165-175.
Pung-amritt P, Poort SR, Vos HL, et al. Compound heterozygosity for one novel and one recurrent mutation in a Thai
patient with severe protein S deficiency. Thromb Haemost.
1999;81(2):189-192.
Sen K, Roy A. Management of neonatal purpura fulminans with severe protein C deficiency. Indian Pediatr.
2006;43(6):542-545.
Tripodi A, Franchi F, Krachmalnicoff A, Mannucci PM.
Asymptomatic homozygous protein C deficiency. Acta
Haematol. 1990;83(3):152-155.

e792S

434. Yamamoto K, Matsushita T, Sugiura I, et al. Homozygous


protein C deficiency: identification of a novel missense
mutation that causes impaired secretion of the mutant
protein C. J Lab Clin Med. 1992;119(6):682-689.
435. Zenciroglu A, Ipek MS, Aydin M, Kara A, Okumus N, Kilic M.
Purpura fulminans in a newborn infant with galactosemia.
Eur J Pediatr. 2010;169(7):903-906.
436. Estells A, Garcia-Plaza I, Das A, et al. Severe inherited
homozygous protein C deficiency in a newborn infant.
Thromb Haemost. 1984;52(1):53-56.
437. Tcheng WY, Dovat S, Gurel Z, Donkin J, Wong WY. Severe
congenital protein C deficiency: description of a new
mutation and prophylactic protein C therapy and in vivo
pharmacokinetics. J Pediatr Hematol Oncol. 2008;30(2):
166-171.
438. Mahasandana C, Suvatte V, Chuansumrit A, et al. Homozygous
protein S deficiency in an infant with purpura fulminans.
J Pediatr. 1990;117(5):750-753.
439. Mahasandana C, Suvatte V, Marlar RA, Manco-Johnson MJ,
Jacobson LJ, Hathaway WE. Neonatal purpura fulminans
associated with homozygous protein S deficiency. Lancet.
1990;335(8680):61-62.
440. Lee MJ, Kim KM, Kim JS, Kim YJ, Lee YJ, Ghim TT.
Long-term survival of a child with homozygous protein C
deficiency successfully treated with living donor liver transplantation. Pediatr Transplant. 2009;13(2):251-254.
441. Andrew M, David M, Adams M, et al. Venous thromboembolic complications (VTE) in children: first analyses
of the Canadian Registry of VTE. Blood. 1994;83(5):
1251-1257.
442. Levy ML, Granville RC, Hart D, Meltzer H. Deep venous
thrombosis in children and adolescents. J Neurosurg.
2004;101(1 suppl):32-37.
443. Nuss R, Hays T, Manco-Johnson M. Childhood thrombosis.
Pediatrics. 1995;96(2 pt 1):291-294.
444. Andrew M, Marzinotto V, Pencharz P, et al. A crosssectional study of catheter-related thrombosis in children
receiving total parenteral nutrition at home. J Pediatr.
1995;126(3):358-363.
445. Krafte-Jacobs B, Sivit CJ, Mejia R, Pollack MM. Catheterrelated thrombosis in critically ill children: comparison of
catheters with and without heparin bonding. J Pediatr. 1995;
126(1):50-54.
446. Massicotte MP, Dix D, Monagle P, Adams M, Andrew M.
Central venous catheter related thrombosis in children: analysis of the Canadian Registry of Venous
Thromboembolic Complications. J Pediatr. 1998;133(6):
770-776.
447. Randolph AG, Cook DJ, Gonzales CA, Andrew M. Benefit
of heparin in central venous and pulmonary artery catheters: a meta-analysis of randomized controlled trials. Chest.
1998;113(1):165-171.
448. Male C, Chait P, Ginsberg JS, et al. Comparison of venography and ultrasound for the diagnosis of asymptomatic deep
vein thrombosis in the upper body in children: results of the
PARKAA study. Prophylactic Antithrombin Replacement in
Kids with ALL treated with Asparaginase. Thromb Haemost.
2002;87(4):593-598.
449. Biss TT, Brando LR, Kahr WH, Chan AK, Williams S.
Clinical features and outcome of pulmonary embolism in
children. Br J Haematol. 2008;142(5):808-818.
450. Derish MT, Smith DW, Frankel LR. Venous catheter
thrombus formation and pulmonary embolism in children.
Pediatr Pulmonol. 1995;20(6):349-354.
451. Kuhle S, Koloshuk B, Marzinotto V, et al. A cross-sectional
study evaluating post-thrombotic syndrome in children.
Thromb Res. 2003;111(4-5):227-233.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

452. Sharathkumar AA, Stanley JC. Management of a child with


renal artery stenosis and homozygous factor V Leiden mutation. J Pediatr Surg. 2008;43(1):e17-e19.
453. Cesaro S, Paris M, Corr R, et al. Successful treatment of
a catheter-related right atrial thrombosis with recombinant
tissue plasminogen activator and heparin. Support Care
Cancer. 2002;10(3):253-255.
454. Knfler R, Dinger J, Kabus M, et al. Thrombolytic therapy
in childrenclinical experiences with recombinant tissueplasminogen activator. Semin Thromb Hemost. 2001;27(2):
169-174.
455. Levitas A, Zucker N, Zalzstein E, Sofer S, Kapelushnik J,
Marks KA. Successful treatment of infective endocarditis
with recombinant tissue plasminogen activator. J Pediatr.
2003;143(5):649-652.
456. Martnez-Tallo E, Campo F, Delgado M, Marfl S, Agulla E.
Thrombus in right atrium in two infants successfully treated
with tissue plasminogen activator. Pediatr Emerg Care.
1997;13(1):37-39.
457. Mathur M, Desai N, Sharma J, Rao SP, Goldman GM.
Management of a large organized intraatrial catheter-tip
thrombus in a child with acquired immunodeficiency syndrome using escalating tissue plasminogen activator infusions. Pediatr Crit Care Med. 2005;6(1):79-82.
458. Ryan CA, Andrew M. Failure of thrombolytic therapy in
four children with extensive thromboses. Am J Dis Child.
1992;146(2):187-193.
459. Schermer E, Streif W, Genser N, Frhwirth M, Trawger R,
Simma B. [Thrombolysis with recombinant tissue-type
plasminogen activator (rt-PA) in 13 children: a case series].
Wien Klin Wochenschr. 2000;112(21):927-933.
460. Van Overmeire B, Van Reempts PJ, Van Acker KJ. Intracardiac thrombus formation with rapidly progressive heart
failure in the neonate: treatment with tissue type plasminogen
activator. Arch Dis Child. 1992;67(4 spec no):443-445.
461. Goldenberg NA, Donadini MP, Kahn SR, et al. Postthrombotic syndrome in children: a systematic review of
frequency of occurrence, validity of outcome measures, and
prognostic factors. Haematologica. 2010;95(11):1952-1959.
462. Leary SE, Harrod VL, de Alarcon PA, Reiss UM. Low-dose
systemic thrombolytic therapy for deep vein thrombosis in
pediatric patients. J Pediatr Hematol Oncol. 2010;32(2):97-102.
463. Oguzkurt L, Ozkan U, Tercan F, Koc Z. Catheter-directed
thrombolysis of acute deep vein thrombosis in the lower
extremity of a child with interrupted inferior vena cava.
Cardiovasc Intervent Radiol. 2007;30(2):332-334.
464. Wicky ST. Acute deep vein thrombosis and thrombolysis.
Tech Vasc Interv Radiol. 2009;12(2):148-153.
465. Brightwell RE, Osman IS. Iliofemoral deep vein thrombosis in childhood; developing a management protocol. Eur J
Vasc Endovasc Surg. 2006;31(6):667-678.
466. Temple M, Williams S, John P, Chait P, Connolly B.
Percutaneous treatment of pediatric thrombosis. Eur J
Radiol. 2005;53(1):14-21.
467. Janssen MC, Wollersheim H, Schultze-Kool LJ, Thien T.
Local and systemic thrombolytic therapy for acute deep
venous thrombosis. Neth J Med. 2005;63(3):81-90.
468. De Blanche LE, Schmitz ML, Johnson CE, Best TH,
Drummond-Webb JJ. Successful surgical management of
a neonate with a saddle pulmonary embolus. Ann Thorac
Surg. 2004;78(1):e1-e2.
469. Poon WL, Luk SH, Yam KY, Lee AC. Mechanical thrombectomy in inferior vena cava thrombosis after caval filter
placement: a report of three cases. Cardiovasc Intervent
Radiol. 2002;25(5):440-443.
470. Putnam JB Jr, Lemmer JH Jr, Rocchini AP, Bove EL.
Embolectomy for acute pulmonary artery occlusion folwww.chestpubs.org

471.
472.
473.
474.
475.

476.

477.
478.
479.
480.
481.
482.

483.
484.
485.
486.

487.

488.

489.
490.

lowing Fontan procedure. Ann Thorac Surg. 1988;45(3):


335-336.
Uflacker R. Interventional therapy for pulmonary embolism.
J Vasc Interv Radiol. 2001;12(2):147-164.
Vincent RN, Dinkins J, Dobbs MC. Mechanical thrombectomy using the AngioJect in a child with congenital heart
disease. Catheter Cardiovasc Interv. 2004;61(2):253-255.
Anton N, Chait P, Chan A, et al. Vena caval filters in children: Preliminary safety and efficacy data. Thromb Haemost.
2001;82(suppl):2227.
Cahn MD, Rohrer MJ, Martella MB, Cutler BS. Long-term
follow-up of Greenfield inferior vena cava filter placement
in children. J Vasc Surg. 2001;34(5):820-825.
Cook A, Shackford S, Osler T, Rogers F, Sartorelli K,
Littenberg B. Use of vena cava filters in pediatric trauma
patients: data from the National Trauma Data Bank.
J Trauma. 2005;59(5):1114-1120.
Haider EA, Rosen JC, Torres C, Valenti DA. Serial repositioning of a Gnther tulip retrievable inferior vena cava
filter in a pediatric patient. Pediatr Radiol. 2005;35(11):
1135-1138.
Bogue CO, John PR, Connolly BL, Rea DJ, Amaral JG.
Symptomatic caval penetration by a Celect inferior vena
cava filter. Pediatr Radiol. 2009;39(10):1110-1113.
Hoppe H, Kaufman JA, Barton RE, et al. Safety of inferior
vena cava filter retrieval in anticoagulated patients. Chest.
2007;132(1):31-36.
Yavuz K, Geyik S, Hoppe H, Kolbeck KJ, Kaufman JA.
Venous thromboembolism after retrieval of inferior vena
cava filters. J Vasc Interv Radiol. 2008;19(4):504-508.
Wiernikowski JT, Athale UH. Thromboembolic complications
in children with cancer. Thromb Res. 2006;118(1):137-152.
Athale U, Cox S, Siciliano S, Chan AK. Thromboembolism
in children with sarcoma. Pediatr Blood Cancer. 2007;49(2):
171-176.
Athale U, Siciliano S, Thabane L, et al. Epidemiology and
clinical risk factors predisposing to thromboembolism in
children with cancer. Pediatr Blood Cancer. 2008;51(6):
792-797.
Athale UH, Nagel K, Khan AA, Chan AK. Thromboembolism
in children with lymphoma. Thromb Res. 2008;122(4):
459-465.
Glaser DW, Medeiros D, Rollins N, Buchanan GR.
Catheter-related thrombosis in children with cancer. J
Pediatr. 2001;138(2):255-259.
Journeycake JM, Buchanan GR. Catheter-related deep
venous thrombosis and other catheter complications in children with cancer. J Clin Oncol. 2006;24(28):4575-4580.
Mitchell L, Andrew M, Hanna K, et al. Trend to efficacy
and safety using antithrombin concentrate in prevention of
thrombosis in children receiving l-asparaginase for acute
lymphoblastic leukemia. Results of the PAARKA study.
Thromb Haemost. 2003;90(2):235-244.
Nowak-Gttl U, Kenet G, Mitchell LG. Thrombosis in
childhood acute lymphoblastic leukaemia: epidemiology,
aetiology, diagnosis, prevention and treatment. Best Pract
Res Clin Haematol. 2009;22(1):103-114.
Tousovska K, Zapletal O, Skotakova J, Bukac J, Sterba J.
Treatment of deep venous thrombosis with low molecular
weight heparin in pediatric cancer patients: safety and efficacy. Blood Coagul Fibrinolysis. 2009;20(7):583-589.
Payne JH, Vora AJ. Thrombosis and acute lymphoblastic
leukaemia. Br J Haematol. 2007;138(4):430-445.
Orsino A, Schneider R, DeVeber G, et al. Childhood acute
myelomonocytic leukemia (AML-M4) presenting as catastrophic antiphospholipid antibody syndrome. J Pediatr
Hematol Oncol. 2004;26(5):327-330.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e793S

491. Athale UH, Chan AK. Thrombosis in children with acute


lymphoblastic leukemia. Part II. Pathogenesis of thrombosis in children with acute lymphoblastic leukemia: effects
of the disease and therapy. Thromb Res. 2003;111(4-5):
199-212.
492. Athale UH, Chan AK. Thrombosis in children with
acute lymphoblastic leukemia Part III. Pathogenesis of
thrombosis in children with acute lymphoblastic leukemia:
effects of host environment. Thromb Res. 2003;111(6):
321-327.
493. Stine KC, Saylors RL, Saccente CS, Becton DL. Treatment
of deep vein thrombosis with enoxaparin in pediatric cancer patients receiving chemotherapy. Clin Appl Thromb
Hemost. 2007;13(2):161-165.
494. Campos LM, Kiss MH, DAmico EA, Silva CA. Antiphospholipid antibodies and antiphospholipid syndrome in
57 children and adolescents with systemic lupus erythematosus. Lupus. 2003;12(11):820-826.
495. Levy DM, Massicotte MP, Harvey E, Hebert D, Silverman
ED. Thromboembolism in paediatric lupus patients. Lupus.
2003;12(10):741-746.
496. Manco-Johnson MJ, Nuss R. Lupus anticoagulant in children with thrombosis. Am J Hematol. 1995;48(4):240-243.
497. Hunt BJ. Pediatric antiphospholipid antibodies and antiphospholipid syndrome. Semin Thromb Hemost. 2008;34(3):
274-281.
498. Kamat AV, DCruz DP, Hunt BJ. Managing antiphospholipid antibodies and antiphospholipid syndrome in children.
Haematologica. 2006;91(12):1674-1680.
499. Male C, Foulon D, Hoogendoorn H, et al. Predictive
value of persistent versus transient antiphospholipid antibody subtypes for the risk of thrombotic events in pediatric patients with systemic lupus erythematosus. Blood.
2005;106(13):4152-4158.
500. Young G, Albisetti M, Bonduel M, et al. Impact of inherited
thrombophilia on venous thromboembolism in children: a
systematic review and meta-analysis of observational studies.
Circulation. 2008;118(13):1373-1382.
501. Raffini L. Thrombophilia in children: who to test, how, when,
and why? Hematology (Am Soc Hematol Educ Program).
2008;2008(1):228-235.
502. Koc Z, Oguzkurt L. Interruption or congenital stenosis of
the inferior vena cava: prevalence, imaging, and clinical
findings. Eur J Radiol. 2007;62(2):257-266.
503. Kondo Y, Koizumi J, Nishibe M, Muto A, Dardik A, Nishibe
T. Deep venous thrombosis caused by congenital absence of
the inferior vena cava: report of a case. Surg Today. 2009;
39(3):231-234.
504. Linnemann B, Schmidt H, Schindewolf M, et al. Etiology
and VTE risk factor distribution in patients with inferior
vena cava thrombosis. Thromb Res. 2008;123(1):72-78.
505. Liu WC, Hung CC, Hwang SJ, Chen HC. Extended septic thrombophlebitis in a patient with duplicated inferior
vena cava: case report and review of literature. Int Angiol.
2009;28(2):156-160.
506. Milio G, Corrado E, Novo S, Licata G, Pinto A. Agenesis
of the renal segment of inferior vena cava associated with
venous stasis. Int Angiol. 2010;29(4):385-388.
507. Nichols JL, Gonzalez SC, Bellino PJ, Bieber EJ. Venous
thrombosis and congenital absence of inferior vena cava in a
patient with menorrhagia and pelvic pain. J Pediatr Adolesc
Gynecol. 2010;23(1):e17-e21.
508. Rose SS, Ali Y, Kumar A, Bekos TJ, Saidi P. Deep venous
thrombosis caused by inferior vena cava atresia and hereditary thrombophilia. Am J Med Sci. 2009;337(1):67-70.
509. Sagban TA, Grotemeyer D, Balzer KM, et al. Surgical treatment for agenesis of the vena cava: a single-centre expee794S

510.

511.

512.
513.

514.

515.
516.
517.
518.
519.

520.
521.

522.

523.

524.
525.
526.

527.

rience in 15 cases. Eur J Vasc Endovasc Surg. 2010;40(2):


241-245.
Brando LR, Williams S, Kahr WH, Ryan C, Temple M,
Chan AK. Exercise-induced deep vein thrombosis of the
upper extremity. 2. A case series in children. Acta Haematol.
2006;115(3-4):221-229.
Brando LR, Williams S, Kahr WH, Ryan C, Temple M,
Chan AK. Exercise-induced deep vein thrombosis of the
upper extremity. 1. Literature review. Acta Haematol.
2006;115(3-4):214-220.
Dhillon RK, Spahr CD. Two cases of upper-extremity
swelling: Paget-Schroetter syndrome and non-Hodgkin lymphoma. Pediatr Emerg Care. 2010;26(4):290-292.
Dintaman J, Watson C, Fox CJ, Hoover N, Roberts S,
Gillespie DL. Case of adolescent with Paget-Schroetter
syndrome and underlying thrombophilia due to an elevated lipoprotein (A). Pediatr Blood Cancer. 2007;49(7):
1036-1038.
Guzzo JL, Chang K, Demos J, Black JH, Freischlag JA.
Preoperative thrombolysis and venoplasty affords no benefit in patency following first rib resection and scalenectomy
for subacute and chronic subclavian vein thrombosis. J Vasc
Surg. 2010;52(3):658-662, discussion 662-663.
Illig KA, Doyle AJ. A comprehensive review of PagetSchroetter syndrome. J Vasc Surg. 2010;51(6):1538-1547.
Landry GJ, Liem TK. Endovascular management of PagetSchroetter syndrome. Vascular. 2007;15(5):290-296.
Molina JE, Hunter DW, Dietz CA. Paget-Schroetter
syndrome treated with thrombolytics and immediate surgery. J Vasc Surg. 2007;45(2):328-334.
Rigberg DA, Gelabert H. The management of thoracic
outlet syndrome in teenaged patients. Ann Vasc Surg.
2009;23(3):335-340.
Roche-Nagle G, Ryan R, Barry M, Brophy D. Effort thrombosis of the upper extremity in a young sportsman: PagetSchroetter syndrome. Br J Sports Med. 2007;41(8):540-541,
discussion 541.
Bendaly EA, Batra AS, Ebenroth ES, Hurwitz RA. Outcome
of cardiac thrombi in infants. Pediatr Cardiol. 2008;29(1):
95-101.
Simon A, Ammann RA, Wiszniewsky G, Bode U, Fleischhack
G, Besuden MM. Taurolidine-citrate lock solution (TauroLock)
significantly reduces CVAD-associated grampositive infections in pediatric cancer patients. BMC Infect Dis. 2008;
8:102.
de Neef M, Heijboer H, van Woensel JB, de Haan RJ. The
efficacy of heparinization in prolonging patency of arterial
and central venous catheters in children: a randomized
double-blind trial. Pediatr Hematol Oncol. 2002;19(8):
553-560.
Smith S, Dawson S, Hennessey R, Andrew M. Maintenance
of the patency of indwelling central venous catheters: is heparin necessary? Am J Pediatr Hematol Oncol. 1991;13(2):
141-143.
Kannan A. Heparinised saline or normal saline? J Perioper
Pract. 2008;18(10):440-441.
Ociepa T, Maloney E, Urasinski T, Sawicki M. Thrombotic
complications of tunneled central lines in children with
malignancy. J Pediatr Hematol Oncol. 2010;32(2):88-92.
Gittins NS, Hunter-Blair YL, Matthews JN, Coulthard MG.
Comparison of alteplase and heparin in maintaining the
patency of paediatric central venous haemodialysis lines:
a randomised controlled trial. Arch Dis Child. 2007;92(6):
499-501.
Dillon PW, Jones GR, Bagnall-Reeb HA, Buckley JD,
Wiener ES, Haase GM; Childrens Oncology Group.
Prophylactic urokinase in the management of long-term
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

venous access devices in children: a Childrens Oncology


Group study. J Clin Oncol. 2004;22(13):2718-2723.
528. Fisher AA, Deffenbaugh C, Poole RL, Garcia M, Kerner
JA Jr. The use of alteplase for restoring patency to occluded
central venous access devices in infants and children. J Infus
Nurs. 2004;27(3):171-174.
529. Ruud E, Holmstrm H, De Lange C, Hogstad EM,
Wesenberg F. Low-dose warfarin for the prevention of
central line-associated thromboses in children with malignanciesa randomized, controlled study. Acta Paediatr.
2006;95(9):1053-1059.
530. Nowak-Gttl U, Mnchow N, Klippel U, et al. The course
of fibrinolytic proteins in children with malignant bone
tumours. Eur J Pediatr. 1999;158(suppl 3):S151-S153.
531. Meister B, Kropshofer G, Klein-Franke A, Strasak AM,
Hager J, Streif W. Comparison of low-molecular-weight
heparin and antithrombin versus antithrombin alone for
the prevention of symptomatic venous thromboembolism in
children with acute lymphoblastic leukemia. Pediatr Blood
Cancer. 2008;50(2):298-303.
532. Dollery CM, Sullivan ID, Bauraind O, Bull C, Milla PJ.
Thrombosis and embolism in long-term central venous access
for parenteral nutrition. Lancet. 1994;344(8929):1043-1045.
533. Mollitt DL, Golladay ES. Complications of TPN catheterinduced vena caval thrombosis in children less than one year
of age. J Pediatr Surg. 1983;18(4):462-467.
534. Moukarzel A, Azancot-Benisty A, Brun P, Vitoux C,
Cezard JP, Navarro J. M-mode and two-dimensional
echocardiography in the routine follow-up of central venous
catheters in children receiving total parenteral nutrition.
JPEN J Parenter Enteral Nutr. 1991;15(5):551-555.
535. Newall F, Barnes C, Savoia H, Campbell J, Monagle P.
Warfarin therapy in children who require long-term total
parenteral nutrition. Pediatrics. 2003;112(5):e386.
536. Ryan JA Jr, Abel RM, Abbott WM, et al. Catheter complications in total parenteral nutrition. A prospective study
of 200 consecutive patients. N Engl J Med. 1974;290(14):
757-761.
537. Wakefield A, Cohen Z, Craig M, et al. Thrombogenicity of
total parenteral nutrition solutions: I. Effect on induction of
monocyte/macrophage procoagulant activity. Gastroenterology.
1989;97(5):1210-1219.
538. Wakefield A, Cohen Z, Rosenthal A, et al. Thrombogenicity
of total parenteral nutrition solutions: II. Effect on induction
of endothelial cell procoagulant activity. Gastroenterology.
1989;97(5):1220-1228.
539. Germanakis I, Sfyridaki C, Papadopoulou E, et al. Stroke
following Glenn anastomosis in a child with inherited thrombophilia. Int J Cardiol. 2006;111(3):464-467.
540. Imanaka K, Takamoto S, Murakami A, Kaneko Y. Right
ventricular thrombosis early after bidirectional Glenn shunt.
Ann Thorac Surg. 1999;68(2):563-565.
541. Kopf GS, Laks H, Stansel HC, Hellenbrand WE, Kleinman
CS, Talner NS. Thirty-year follow-up of superior vena cavapulmonary artery (Glenn) shunts. J Thorac Cardiovasc Surg.
1990;100(5):662--671.
542. Koutlas TC, Harrison JK, Bashore TM, OLauglin MP, Tripp
ME, Gaynor JW. Late conduit occlusion after modified
Fontan procedure with classic Glenn shunt. Ann Thorac Surg.
1996;62(1):258-262.
543. Pennington DG, Nouri S, Ho J, et al. Glenn shunt: longterm results and current role in congenital heart operations.
Ann Thorac Surg. 1981;31(6):532-539.
544. Sreeram N, Emmel M, Trieschmann U, et al. Reopening
acutely occluded cavopulmonary connections in infants
and children. Interact Cardiovasc Thorac Surg. 2010;10(3):
383-388.
www.chestpubs.org

545. Ono M, Boethig D, Goerler H, Lange M, Westhoff-Bleck M,


Breymann T. Clinical outcome of patients 20 years after
Fontan operationeffect of fenestration on late morbidity.
Eur J Cardiothorac Surg. 2006;30(6):923-929.
546. Tweddell JS, Nersesian M, Mussatto KA, et al. Fontan palliation in the modern era: factors impacting mortality and
morbidity. Ann Thorac Surg. 2009;88(4):1291-1299.
547. Monagle P, Cochrane A, McCrindle B, Benson L, Williams
W, Andrew M. Thromboembolic complications after Fontan
proceduresthe role of prophylactic anticoagulation.
J Thorac Cardiovasc Surg. 1998;115(3):493-498.
548. Monagle P, Karl TR. Thromboembolic problems after the
Fontan operation. Semin Thorac Cardiovasc Surg Pediatr
Card Surg Annu. 2002;5:36-47.
549. Mosquera VX, Marini M, Portela F, Cao I. Late complication of classic Fontan operation: giant right atrial thrombus
and massive pulmonary thromboembolism. J Card Surg.
2008;23(6):776-778.
550. Balling G, Vogt M, Kaemmerer H, Eicken A, Meisner H,
Hess J. Intracardiac thrombus formation after the Fontan
operation. J Thorac Cardiovasc Surg. 2000;119(4 pt 1):
745-752.
551. Fyfe DA, Kline CH, Sade RM, Gillette PC. Transesophageal
echocardiography detects thrombus formation not identified
by transthoracic echocardiography after the Fontan operation. J Am Coll Cardiol. 1991;18(7):1733-1737.
552. Stmper O, Sutherland GR, Geuskens R, Roelandt JR,
Bos E, Hess J. Transesophageal echocardiography in evaluation and management after a Fontan procedure. J Am Coll
Cardiol. 1991;17(5):1152-1160.
553. Takawira F, Ayer JG, Onikul E, et al. Evaluation of the
extracardiac conduit modification of the Fontan operation
for thrombus formation using magnetic resonance imaging.
Heart Lung Circ. 2008;17(5):407-410.
554. Kudumula V, Mathur S, BuLock F. Successful thrombolysis
of massive intracardiac thrombus in atriopulmonary Fontan
circulation. Cardiol Young. 2010;20(4):443-444.
555. Rauch R, Sieverding L, Hofbeck M. Thrombosis of an extracardiac Fontan tunnel: combined treatment of thrombolysis
and stenting. Catheter Cardiovasc Interv. 2009;74(6):917-919.
556. Kaulitz R, Ziemer G, Rauch R, et al. Prophylaxis of thromboembolic complications after the Fontan operation (total
cavopulmonary anastomosis). J Thorac Cardiovasc Surg.
2005;129(3):569-575.
557. Seipelt RG, Franke A, Vazquez-Jimenez JF, et al. Thromboembolic complications after Fontan procedures: comparison of different therapeutic approaches. Ann Thorac Surg.
2002;74(2):556-562.
558. Khairy P, Fernandes SM, Mayer JE Jr, et al. Long-term survival, modes of death, and predictors of mortality in patients
with Fontan surgery. Circulation. 2008;117(1):85-92.
559. Robbers-Visser D, Miedema M, Nijveld A, et al. Results
of staged total cavopulmonary connection for functionally
univentricular hearts; comparison of intra-atrial lateral
tunnel and extracardiac conduit. Eur J Cardiothorac Surg.
2010;37(4):934-941.
560. Monagle P, Cochrane A, Roberts R, et al; Fontan
Anticoagulation Study Group. A multicentre randomized
trial comparing heparin/warfarin versus acetylsalicylic acid
as primary thromboprophylaxis for two years after Fontan
procedure in children. J Am Coll Cardiol. 2011;58(6):645-651.
561. Bakkaloglu SA, Keefer MS, Schroeder S, Stanley P, Harrell
DS, Reiff A. Experience with multiple stent implantations
in primary antiphospholipid syndrome in childhood: a case
report. Clin Exp Rheumatol. 2009;27(4):664-667.
562. Chang HL, Patel VI, Brewster DC, Masiakos PT. Endovascular stenting of a penetrating axillary artery injury
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e795S

563.

564.

565.
566.
567.

568.

569.

570.

571.

572.
573.

574.
575.
576.
577.
578.
579.
580.

581.

in a 14-year-old with 1-year follow-up. J Pediatr Surg.


2009;44(1):294-297.
Gunabushanam V, Mishra N, Calderin J, Glick R, Rosca
M, Krishnasastry K. Endovascular stenting of blunt thoracic aortic injury in an 11-year-old. J Pediatr Surg. 2010;
45(3):E15-E18.
Holzer R, Qureshi S, Ghasemi A, et al. Stenting of aortic
coarctation: acute, intermediate, and long-term results
of a prospective multi-institutional registryCongenital
Cardiovascular Interventional Study Consortium (CCISC).
Catheter Cardiovasc Interv. 2010;76(4):553-563.
Lai YJ, Chang FC, Lin CJ, Hsieh TC, Wang KL. Endovascular
therapy in pediatric intracranial carotid artery dissection.
Pediatr Neurol. 2010;42(4):291-294.
Lv X, Jiang C, Li Y, Yang X, Wu Z. Endovascular treatment for pediatric intracranial aneurysms. Neuroradiology.
2009;51(11):749-754.
Mazzei A, Centonze A, Stranieri G, Placida G, Siani A,
Baldassarre E. Arterial vascular injuries of the upper arm in
children: saphenous vein graft or endovascular treatment? J
Pediatr Surg. 2010;45(4):850-851.
Oguzkurt L, Tercan F, Sener M. Successful endovascular treatment of iliac vein compression (May-Thurner)
syndrome in a pediatric patient. Cardiovasc Intervent
Radiol. 2006;29(3):446-449.
Patnaik AN, Srinivas B, Rao DS. Endovascular stenting for
native coarctation in older children and adolescents using
adult self-expanding (Nitinol) iliac stents. Indian Heart J.
2009;61(4):353-357.
Radanovi B, Caci Z, Perkov D, Smiljani R, Cori SR,
Ilakovac K. Endovascular therapy of renovascular hypertension in children: single center analysis. Eur J Pediatr Surg.
2009;19(3):135-140.
Sachdeva R, Seib PM, Burns SA, Fontenot EE, Frazier EA.
Stenting for superior vena cava obstruction in pediatric heart
transplant recipients. Catheter Cardiovasc Interv. 2007;
70(6):888-892.
Tullus K, Brennan E, Hamilton G, et al. Renovascular hypertension in children. Lancet. 2008;371(9622):1453-1463.
Zahn EM, Lima VC, Benson LN, Freedom RM. Use of
endovascular stents to increase pulmonary blood flow in pulmonary atresia with ventricular septal defect. Am J Cardiol.
1992;70(3):411-412.
Ashwath R, Gruenstein D, Siwik E. Percutaneous stent
placement in children weighing less than 10 kilograms.
Pediatr Cardiol. 2008;29(3):562-567.
John JB, Cron SG, Kung GC, Mott AR. Intracardiac thrombi
in pediatric patients: presentation profiles and clinical outcomes. Pediatr Cardiol. 2007;28(3):213-220.
Yilmazer MM, Guven B, Tavli V. An unusual form of intracardiac thrombosis and fibrinolytic process in a child with
dilated cardiomyopathy. Acta Cardiol. 2010;65(3):341-343.
Hsu DT, Addonizio LJ, Hordof AJ, Gersony WM. Acute
pulmonary embolism in pediatric patients awaiting heart
transplantation. J Am Coll Cardiol. 1991;17(7):1621-1625.
McCrindle BW, Karamlou T, Wong H, et al. Presentation,
management and outcomes of thrombosis for children with
cardiomyopathy. Can J Cardiol. 2006;22(8):685-690.
Baker DW, Wright RF. Management of heart failure. IV.
Anticoagulation for patients with heart failure due to left
ventricular systolic dysfunction. JAMA. 1994;272(20):1614-1618.
Adatia I, Kothari SS, Feinstein JA. Pulmonary hypertension associated with congenital heart disease: pulmonary
vascular disease: the global perspective. Chest. 2010;137
(suppl 6):52S-61S.
Hawkins A, Tulloh R. Treatment of pediatric pulmonary
hypertension. Vasc Health Risk Manag. 2009;5(2):509-524.

e796S

582. Rashid A, Ivy D. Severe paediatric pulmonary hypertension:


new management strategies. Arch Dis Child. 2005;90(1):
92-98.
583. Ivy D. Diagnosis and treatment of severe pediatric pulmonary hypertension. Cardiol Rev. 2001;9(4):227-237.
584. Krishnan U. Diagnosis and management of primary pulmonary hypertension. Indian J Pediatr. 2000;67(3 suppl):
S41-S45.
585. Badesch DB, Abman SH, Ahearn GS, et al; American
College of Chest Physicians. Medical therapy for pulmonary
arterial hypertension: ACCP evidence-based clinical practice guidelines. Chest. 2004;126(1 suppl):35S-62S.
586. Barst RJ. Recent advances in the treatment of pediatric
pulmonary artery hypertension. Pediatr Clin North Am.
1999;46(2):331-345.
587. Rosenzweig EB, Barst RJ. Idiopathic pulmonary arterial
hypertension in children. Curr Opin Pediatr. 2005;17(3):
372-380.
588. Yankah AC, Alexi-Meskhishvili V, Weng Y, Berger F,
Lange P, Hetzer R. Performance of aortic and pulmonary
homografts in the right ventricular outflow tract in children.
J Heart Valve Dis. 1995;4(4):392-395.
589. Turrentine MW, Ruzmetov M, Vijay P, Bills RG, Brown JW.
Biological versus mechanical aortic valve replacement in
children. Ann Thorac Surg. 2001;71(5 suppl):S356-S360.
590. Caldarone CA, Raghuveer G, Hills CB, et al. Longterm survival after mitral valve replacement in children
aged ,5 years: a multi-institutional study. Circulation.
2001;104(12)(suppl 1):I143-I147.
591. Kadoba K, Jonas RA, Mayer JE, Castaneda AR. Mitral valve
replacement in the first year of life. J Thorac Cardiovasc
Surg. 1990;100(5):762-768.
592. Milano A, Vouh PR, Baillot-Vernant F, et al. Late results
after left-sided cardiac valve replacement in children.
J Thorac Cardiovasc Surg. 1986;92(2):218-225.
593. Spevak PJ, Freed MD, Castaneda AR, Norwood WI, Pollack
P. Valve replacement in children less than 5 years of age.
J Am Coll Cardiol. 1986;8(4):901-908.
594. Weinstein GS, Mavroudis C, Ebert PA. Preliminary
experience with aspirin for anticoagulation in children with
prosthetic cardiac valves. Ann Thorac Surg. 1982;33(6):
549-553.
595. Whitlock RP, Sun JC, Fremes SE, Rubens FD, Teoh KH.
Antithrombotic and thrombolytic therapy for valvular disease: antithrombotic therapy and prevention of thrombosis,
9th ed: American College of Chest Physicians evidencebased clinical practice guidelines. Chest. 2012;141(2)(suppl):
e576S-e600S.
596. Alsoufi B, Manlhiot C, McCrindle BW, et al. Results after
mitral valve replacement with mechanical prostheses in
young children. J Thorac Cardiovasc Surg. 2010;139(5):
1189-1196.
597. Serra AJ, McNicholas KW, Olivier HF Jr, Boe SL, Lemole
GM. The choice of anticoagulation in pediatric patients with
the St. Jude Medical valve prostheses. J Cardiovasc Surg
(Torino). 1987;28(5):588-591.
598. Abid F, Abid A, Fekih M, Zaouali RM, Ben Ismail M. Aortic
valve replacement in children under 16 years of age with
congenital or rheumatic valvular disease. A study of 64 cases.
J Cardiovasc Surg (Torino). 1992;33(3):265-271.
599. Borkon AM, Soule L, Reitz BA, Gott VL, Gardner TJ.
Five year follow-up after valve replacement with the St. Jude
Medical valve in infants and children. Circulation. 1986;
74(3 pt 2):I110-I115.
600. Khitin LM, Sade RM, Bradley SM, Crawford FA Jr,
Widener CE, Stroud MR. Prevention of thrombosis and
embolism in children and adolescents with mechanical
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

601.
602.

603.
604.

605.

606.
607.
608.
609.
610.
611.
612.

613.

614.
615.

616.

617.
618.
619.

valve prostheses: warfarin versus antiplatelet agents. J Heart


Valve Dis. 2006;15(3):394-399, discussion 399.
Robbins RC, Bowman FO Jr, Malm JR. Cardiac valve
replacement in children: a twenty-year series. Ann Thorac
Surg. 1988;45(1):56-61.
Schaffer MS, Clarke DR, Campbell DN, Madigan CK,
Wiggins JW Jr, Wolfe RR. The St. Jude Medical cardiac
valve in infants and children: role of anticoagulant therapy.
J Am Coll Cardiol. 1987;9(1):235-239.
Verrier ED, Tranbaugh RF, Soifer SJ, Yee ES, Turley K,
Ebert PA. Aspirin anticoagulation in children with mechanical
aortic valves. J Thorac Cardiovasc Surg. 1986;92(6):1013-1020.
Akhtar RP, Abid AR, Zafar H, Sheikh SS, Cheema MA,
Khan JS. Prosthetic valve replacement in adolescents with
rheumatic heart disease. Asian Cardiovasc Thorac Ann.
2007;15(6):476-481.
Alexiou C, Galogavrou M, Chen Q, et al. Mitral valve
replacement with mechanical prostheses in children:
improved operative risk and survival. Eur J Cardiothorac
Surg. 2001;20(1):105-113.
Bradley LM, Midgley FM, Watson DC, Getson PR, Scott LP III.
Anticoagulation therapy in children with mechanical prosthetic cardiac valves. Am J Cardiol. 1985;56(8):533-535.
Bradley SM, Sade RM, Crawford FA Jr, Stroud MR.
Anticoagulation in children with mechanical valve prostheses. Ann Thorac Surg. 1997;64(1):30-34, discussion 35-36.
Champsaur G, Robin J, Tronc F, et al. Mechanical valve in
aortic position is a valid option in children and adolescents.
Eur J Cardiothorac Surg. 1997;11(1):117-122.
Kojori F, Chen R, Caldarone CA, et al. Outcomes of mitral
valve replacement in children: a competing-risks analysis.
J Thorac Cardiovasc Surg. 2004;128(5):703-709.
Rao PS, Solymar L, Mardini MK, Fawzy ME, Guinn G.
Anticoagulant therapy in children with prosthetic valves.
Ann Thorac Surg. 1989;47(4):589-592.
Schaff HV, Danielson GK. Current status of valve replacement in children. Cardiovasc Clin. 1986;16(2):427-436.
Stewart S, Cianciotta D, Alexson C, Manning J. The longterm risk of warfarin sodium therapy and the incidence of
thromboembolism in children after prosthetic cardiac valve
replacement. J Thorac Cardiovasc Surg. 1987;93(4):551-554.
Williams JB, Karp RB, Kirklin JW, et al. Considerations in
selection and management of patients undergoing valve
replacement with glutaraldehyde-fixed porcine bioprotheses. Ann Thorac Surg. 1980;30(3):247-258.
Sachweh JS, Tiete AR, Mhler EG, et al. Mechanical aortic
and mitral valve replacement in infants and children. Thorac
Cardiovasc Surg. 2007;55(3):156-162.
Dziuban EJ, Teitelbaum DH, Bakhtyar A, et al. Mesenteric
pseudoaneurysm and cerebral stroke as sequelae of infective endocarditis in an adolescent. J Pediatr Surg. 2008;
43(10):1923-1927.
Lertsapcharoen
P,
Khongphatthanayothin
A,
Chotivittayatarakorn P, Thisyakorn C, Pathmanand C,
Sueblinvong V. Infective endocarditis in pediatric patients:
an eighteen-year experience from King Chulalongkorn
Memorial Hospital. J Med Assoc Thai. 2005;88(suppl 4):
S12-S16.
Sexton DJ, Spelman D. Current best practices and guidelines. Assessment and management of complications in
infective endocarditis. Cardiol Clin. 2003;21(2):273-282.
Niwa K, Nakazawa M, Tateno S, Yoshinaga M, Terai M.
Infective endocarditis in congenital heart disease: Japanese
national collaboration study. Heart. 2005;91(6):795-800.
Delmo Walter EM, Musci M, Nagdyman N, Hbler M,
Berger F, Hetzer R. Mitral valve repair for infective endocarditis in children. Ann Thorac Surg. 2007;84(6):2059-2065.

www.chestpubs.org

620. Zhu HS, Yao PY, Zheng JH, Pezzella AT. Early surgical
intervention for infective endocarditis. Asian Cardiovasc
Thorac Ann. 2002;10(4):298-301.
621. Fynn-Thompson F, Almond C. Pediatric ventricular assist
devices. Pediatr Cardiol. 2007;28(2):149-155.
622. Hetzer R, Alexi-Meskishvili V, Weng Y, et al. Mechanical
cardiac support in the young with the Berlin Heart EXCOR
pulsatile ventricular assist device: 15 years experience.
Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu.
2006:99-108.
623. Potapov EV, Stiller B, Hetzer R. Ventricular assist devices
in children: current achievements and future perspectives.
Pediatr Transplant. 2007;11(3):241-255.
624. Rockett SR, Bryant JC, Morrow WR, et al. Preliminary
single center North American experience with the Berlin
Heart pediatric EXCOR device. ASAIO J. 2008;54(5):
479-482.
625. Russo P, Wheeler A, Russo J, Tobias JD. Use of a ventricular assist device as a bridge to transplantation in a patient
with single ventricle physiology and total cavopulmonary
anastomosis. Paediatr Anaesth. 2008;18(4):320-324.
626. Stiller B, Weng Y, Hbler M, et al. Pneumatic pulsatile ventricular assist devices in children under 1 year of age. Eur J
Cardiothorac Surg. 2005;28(2):234-239.
627. Araba FA, Tsau PH, Smith RG, et al. Pediatric bridge
to heart transplantation: application of the Berlin Heart,
Medos and Thoratec ventricular assist devices. J Heart Lung
Transplant. 2006;25(1):16-21.
628. Blume ED, Naftel DC, Bastardi HJ, Duncan BW,
Kirklin JK, Webber SA; Pediatric Heart Transplant
Study Investigators. Outcomes of children bridged to
heart transplantation with ventricular assist devices:
a multi-institutional study. Circulation. 2006;113(19):
2313-2319.
629. Duncan BW, Bohn DJ, Atz AM, French JW, Laussen PC,
Wessel DL. Mechanical circulatory support for the treatment of children with acute fulminant myocarditis. J Thorac
Cardiovasc Surg. 2001;122(3):440-448.
630. Fraser CD Jr, Carberry KE, Owens WR, et al. Preliminary
experience with the MicroMed DeBakey pediatric ventricular assist device. Semin Thorac Cardiovasc Surg Pediatr
Card Surg Annu. 2006:109-114.
631. Hetzer R, Loebe M, Potapov EV, et al. Circulatory support
with pneumatic paracorporeal ventricular assist device in
infants and children. Ann Thorac Surg. 1998;66(5):1498-1506.
632. Kaczmarek I, Sachweh J, Groetzner J, et al. Mechanical
circulatory support in pediatric patients with the MEDOS
assist device. ASAIO J. 2005;51(5):498-500.
633. Levi D, Marelli D, Plunkett M, et al. Use of assist devices and
ECMO to bridge pediatric patients with cardiomyopathy to
transplantation. J Heart Lung Transplant. 2002;21(7):760-770.
634. Minami K, Knyphausen E, Suzuki R, et al. Mechanical ventricular circulatory support in children; Bad Oeynhausen
experience. Ann Thorac Cardiovasc Surg. 2005;11(5):
307-312.
635. Morales DL, Dibardino DJ, McKenzie ED, et al. Lessons
learned from the first application of the DeBakey VAD
Child: an intracorporeal ventricular assist device for children. J Heart Lung Transplant. 2005;24(3):331-337.
636. Reinhartz O, Hill JD, Al-Khaldi A, Pelletier MP, Robbins
RC, Farrar DJ. Thoratec ventricular assist devices in
pediatric patients: update on clinical results. ASAIO J.
2005;51(5):501-503.
637. Reinhartz O, Keith FM, El-Banayosy A, et al. Multicenter
experience with the thoratec ventricular assist device
in children and adolescents. J Heart Lung Transplant.
2001;20(4):439-448.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e797S

638. Schmid C, Debus V, Gogarten W, et al. Pediatric assist with


the Medos and Excor systems in small children. ASAIO J.
2006;52(5):505-508.
639. Schmid C, Tjan T, Etz C, et al. The excor device-revival
of an old system with excellent results. Thorac Cardiovasc
Surg. 2006;54(6):393-399.
640. Sharma MS, Webber SA, Morell VO, et al. Ventricular assist
device support in children and adolescents as a bridge to heart
transplantation. Ann Thorac Surg. 2006;82(3):926-932.
641. Studer MA, Kennedy CE, Dreyer WJ, et al. An alternative
treatment strategy for pump thrombus in the DeBakey VAD
Child: use of clopidogrel as a thrombolytic agent. J Heart
Lung Transplant. 2006;25(7):857-861.
642. Undar A, McKenzie ED, McGarry MC, et al. Outcomes
of congenital heart surgery patients after extracorporeal
life support at Texas Childrens Hospital. Artif Organs.
2004;28(10):963-966.
643. Ramage IJ, Bailie A, Tyerman KS, McColl JH, Pollard SG,
Fitzpatrick MM. Vascular access survival in children and
young adults receiving long-term hemodialysis. Am J Kidney
Dis. 2005;45(4):708-714.
644. Chand DH, Valentini RP, Kamil ES. Hemodialysis vascular access options in pediatrics: considerations for
patients and practitioners. Pediatr Nephrol. 2009;24(6):
1121-1128.
645. Sheth RD, Brandt ML, Brewer ED, Nuchtern JG, Kale AS,
Goldstein SL. Permanent hemodialysis vascular access
survival in children and adolescents with end-stage renal
disease. Kidney Int. 2002;62(5):1864-1869.
646. Sharathkumar A, Hirschl R, Pipe S, Crandell C, Adams B,
Lin JJ. Primary thromboprophylaxis with heparins for arteriovenous fistula failure in pediatric patients. J Vasc Access.
2007;8(4):235-244.
647. Molitor B, Klingel R, Hafner G. [Monitoring of the heparin therapy during acute haemodialysis]. Hamostaseologie.
2005;25(3):272-278., quiz 279-280.
648. Ozen S, Saati U, Bakkalo lu A, Uyumaz H, Kavuku S.
Tight heparin regimen for haemodialysis in children. Int
Urol Nephrol. 1993;25(5):499-501.
649. Bianchetti MG, Speck S, Mller R, Oetliker OH. Simple
coagulation prophylaxis using low-molecular heparin enoxaparin in pediatric hemodialysis [in German]. Schweiz
Rundsch Med Prax. 1990;79(23):730-731.
650. Van Biljon I, Van Damme-Lombaerts R, Demol A, Van Geet C,
Proesmans W, Arnout J. Low molecular weight heparin for
anticoagulation during haemodialysis in childrena preliminary study. Eur J Pediatr. 1996;155(1):70.
651. Kreuzer M, Bonzel KE, Bscher R, Offner G, Ehrich JH,
Pape L. Regional citrate anticoagulation is safe in intermittent high-flux haemodialysis treatment of children and
adolescents with an increased risk of bleeding. Nephrol Dial
Transplant. 2010;25(10):3337-3342.
652. Moritz ML, Vats A, Ellis D. Systemic anticoagulation and
bleeding in children with hemodialysis catheters. Pediatr
Nephrol. 2003;18(1):68-70.
653. Newburger JW, Takahashi M, Gerber MA, et al; Committee
on Rheumatic Fever, Endocarditis and Kawasaki Disease;
Council on Cardiovascular Disease in the Young; American
Heart Association; American Academy of Pediatrics. Diagnosis,
treatment, and long-term management of Kawasaki disease:
a statement for health professionals from the Committee
on Rheumatic Fever, Endocarditis and Kawasaki Disease,
Council on Cardiovascular Disease in the Young, American
Heart Association. Circulation. 2004;110(17):2747-2771.
654. Catella-Lawson F, Reilly MP, Kapoor SC, et al.
Cyclooxygenase inhibitors and the antiplatelet effects of
aspirin. N Engl J Med. 2001;345(25):1809-1817.
e798S

655. Newburger JW, Takahashi M, Burns JC, et al. The treatment of Kawasaki syndrome with intravenous gamma globulin. N Engl J Med. 1986;315(6):341-347.
656. Ichihashi K, Shiraishi H, Momoi M. Prediction of responsiveness or non-responsiveness to treatment of acute
Kawasaki disease using 1 gram per kilogram of immunoglobulinan effective and cost-saving schedule of therapy.
Cardiol Young. 2009;19(3):224-227.
657. Egami K, Muta H, Ishii M, et al. Prediction of resistance
to intravenous immunoglobulin treatment in patients with
Kawasaki disease. J Pediatr. 2006;149(2):237-240.
658. Newburger JW, Sleeper LA, McCrindle BW, et al; Pediatric
Heart Network Investigators. Randomized trial of pulsed
corticosteroid therapy for primary treatment of Kawasaki
disease. N Engl J Med. 2007;356(7):663-675.
659. Suda K, Kudo Y, Higaki T, et al. Multicenter and retrospective case study of warfarin and aspirin combination
therapy in patients with giant coronary aneurysms caused by
Kawasaki disease. Circ J. 2009;73(7):1319-1323.
660. Manlhiot C, Brando LR, Somji Z, et al. Long-term anticoagulation in Kawasaki disease: Initial use of low molecular weight
heparin is a viable option for patients with severe coronary
artery abnormalities. Pediatr Cardiol. 2010;31(6):834-842.
661. Rowley AH, Shulman ST. Pathogenesis and management of Kawasaki disease. Expert Rev Anti Infect Ther.
2010;8(2):197-203.
662. Pruetz JD, Takahashi M, Reemtsen BL, Starnes VA. A
novel surgical approach to left main coronary artery giant
aneurysm thrombosis in a child with a history of Kawasaki
disease. J Thorac Cardiovasc Surg. 2009;137(4):1030-1032.
663. Yeu BK, Menahem S, Goldstein J. Giant coronary artery
aneurysms in Kawasaki diseasethe need for coronary
artery bypass. Heart Lung Circ. 2008;17(5):404-406.
664. JCS Joint Working Group. Guidelines for diagnosis and
management of cardiovascular sequelae in Kawasaki disease
(JCS 2008)digest version. Circ J. 2010;74(9):1989-2020.
665. DeVeber G. In pursuit of evidence-based treatments for
paediatric stroke: the UK and Chest guidelines. Lancet
Neurol. 2005;4(7):432-436.
666. Barnes C, Newall F, Furmedge J, Mackay M, Monagle P.
Cerebral sinus venous thrombosis in children. J Paediatr
Child Health. 2004;40(1-2):53-55.
667. Huisman TA, Holzmann D, Martin E, Willi UV.
Cerebral venous thrombosis in childhood. Eur Radiol.
2001;11(9):1760-1765.
668. Sbire G, Tabarki B, Saunders DE, et al. Cerebral venous
sinus thrombosis in children: risk factors, presentation, diagnosis and outcome. Brain. 2005;128(pt 3):477-489.
669. Macchi PJ, Grossman RI, Gomori JM, Goldberg HI,
Zimmerman RA, Bilaniuk LT. High field MR imaging
of cerebral venous thrombosis. J Comput Assist Tomogr.
1986;10(1):10-15.
670. Carpenter J, Tsuchida T. Cerebral sinovenous thrombosis in
children. Curr Neurol Neurosci Rep. 2007;7(2):139-146.
671. Roach ES, Golomb MR, Adams R, et al; American Heart
Association Stroke Council; Council on Cardiovascular
Disease in the Young. Management of stroke in infants
and children: a scientific statement from a Special Writing
Group of the American Heart Association Stroke Council
and the Council on Cardiovascular Disease in the Young.
Stroke. 2008;39(9):2644-2691.
672. de Bruijn SF, Stam J. Randomized, placebo-controlled trial
of anticoagulant treatment with low-molecular-weight heparin for cerebral sinus thrombosis. Stroke. 1999;30(3):484-488.
673. Einhupl KM, Villringer A, Meister W, et al. Heparin treatment in sinus venous thrombosis. Lancet. 1991;338(8767):
597-600.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

674. Maiti B, Chakrabarti I. Study on cerebral venous thrombosis


with special reference to efficacy of heparin. J Neurol Sci.
1997;150(suppl 1):S147.
675. Nagaraja D, Rao B, Taly A, et al. Randomized controlled
trial of heparin in puerperal cerebral venous/sinus thrombosis. Nimhans J. 1995;13:111-115.
676. Stam J, De Bruijn SF, DeVeber G. Anticoagulation for
cerebral sinus thrombosis. Cochrane Database Syst Rev.
2002;(4):CD002005.
677. Einhupl K, Bousser MG, de Bruijn SF, et al. EFNS guideline on the treatment of cerebral venous and sinus thrombosis. Eur J Neurol. 2006;13(6):553-559.
678. deVeber G, Monagle P, Chan A, et al. Prothrombotic disorders in infants and children with cerebral thromboembolism. Arch Neurol. 1998;55(12):1539-1543.
679. Bonduel M, Sciuccati G, Hepner M, et al. Arterial ischemic
stroke and cerebral venous thrombosis in children: a
12-year Argentinean registry. Acta Haematol. 2006;115(3-4):
180-185.
680. De Schryver EL, Blom I, Braun KP, et al. Long-term prognosis of cerebral venous sinus thrombosis in childhood. Dev
Med Child Neurol. 2004;46(8):514-519.
681 . deVeber G , Chan A, Monagle P, et al. Anticoagulation
therapy in pediatric patients with sinovenous thrombosis: a
cohort study. Arch Neurol. 1998;55(12):1533-1537.
682. Fluss J, Geary D, deVeber G. Cerebral sinovenous thrombosis and idiopathic nephrotic syndrome in childhood: report
of four new cases and review of the literature. Eur J Pediatr.
2006;165(10):709-716.
683. Holzmann D, Huisman TA, Linder TE. Lateral dural
sinus thrombosis in childhood. Laryngoscope. 1999;109(4):
645-651.
684. Johnson MC, Parkerson N, Ward S, de Alarcon PA.
Pediatric sinovenous thrombosis. J Pediatr Hematol Oncol.
2003;25(4):312-315.
685. Kenet G, Kirkham K, Niederstadt T, et al.; European
Thromboses Study Group. Risk factors for recurrent venous
thromboembolism in the European collaborative paediatric database on cerebral venous thrombosis: a multicentre
cohort study. Lancet Neurol. 2007;6(7):595-603.
686. Kenet G, Waldman D, Lubetsky A, et al. Paediatric cerebral sinus vein thrombosis. A multi-center, case-controlled
study. Thromb Haemost. 2004;92(4):713-718.
687. Uziel Y, Laxer RM, Blaser S, Andrew M, Schneider R,
Silverman ED. Cerebral vein thrombosis in childhood
systemic lupus erythematosus. J Pediatr. 1995;126(5 Pt 1):
722-727.
688. Kenet G, Ltkhoff LK, Albisetti M, et al. Impact of thrombophilia on risk of arterial ischemic stroke or cerebral
sinovenous thrombosis in neonates and children: a systematic review and meta-analysis of observational studies.
Circulation. 2010;121(16):1838-1847.
689. Canho P, Falco F, Ferro JM. Thrombolytics for cerebral
sinus thrombosis: a systematic review. Cerebrovasc Dis.
2003;15(3):159-166.
690. Griesemer DA, Theodorou AA, Berg RA, Spera TD. Local
fibrinolysis in cerebral venous thrombosis. Pediatr Neurol.
1994;10(1):78-80.
691. Liebetrau M, Mayer TE, Bruning R, Opherk C, Hamann GF.
Intra-arterial thrombolysis of complete deep cerebral venous
thrombosis. Neurology. 2004;63(12):2444-2445.
692. Soleau SW, Schmidt R, Stevens S, Osborn A, MacDonald JD.
Extensive experience with dural sinus thrombosis.
Neurosurgery. 2003;52(3):534-544, discussion 542-544.
693. Wasay M, Bakshi R, Kojan S, Bobustuc G, Dubey N,
Unwin DH. Nonrandomized comparison of local urokinase thrombolysis versus systemic heparin anticoagulation
www.chestpubs.org

694.

695.

696.
697.
698.

699.

700.
701.

702.
703.
704.

705.
706.

707.
708.

709.
710.
711.

for superior sagittal sinus thrombosis. Stroke. 2001;32(10):


2310-2317.
Chahlavi A, Steinmetz MP, Masaryk TJ, Rasmussen PA. A
transcranial approach for direct mechanical thrombectomy
of dural sinus thrombosis. Report of two cases. J Neurosurg.
2004;101(2):347-351.
Kulcsr Z, Marosfoi M, Berentei Z, Szikora I. Continuous
thrombolysis and repeated thrombectomy with the Penumbra
System in a child with hemorrhagic sinus thrombosis: technical note. Acta Neurochir (Wien). 2010;152(5):911-916.
Samuel J, Fernandes CM. Lateral sinus thrombosis (a review
of 45 cases). J Laryngol Otol. 1987;101(12):1227-1229.
Ciccone A, Canho P, Falco F, Ferro JM, Sterzi R.
Thrombolysis for cerebral vein and dural sinus thrombosis.
Cochrane Database Syst Rev. 2004;(1):CD003693.
Keller E, Pangalu A, Fandino J, Kn D, Yonekawa Y.
Decompressive craniectomy in severe cerebral venous
and dural sinus thrombosis. Acta Neurochir Suppl (Wien).
2005;94:177-183.
Stefini R, Latronico N, Cornali C, Rasulo F, Bollati A.
Emergent decompressive craniectomy in patients with fixed
dilated pupils due to cerebral venous and dural sinus thrombosis: report of three cases. Neurosurgery. 1999;45(3):626629, discussion 629-630.
Fullerton HJ, Wu YW, Zhao S, Johnston SC. Risk of stroke
in children: ethnic and gender disparities. Neurology.
2003;61(2):189-194.
Giroud M, Lemesle M, Gouyon JB, Nivelon JL, Milan C,
Dumas R. Cerebrovascular disease in children under 16
years of age in the city of Dijon, France: a study of incidence
and clinical features from 1985 to 1993. J Clin Epidemiol.
1995;48(11):1343-1348.
Zahuranec DB, Brown DL, Lisabeth LD, Morgenstern LB.
Is it time for a large, collaborative study of pediatric stroke?
Stroke. 2005;36(9):1825-1829.
Everts R, Pavlovic J, Kaufmann F, et al. Cognitive functioning, behavior, and quality of life after stroke in childhood. Child Neuropsychol. 2008;14(4):323-338.
Goldenberg NA, Bernard TJ, Fullerton HJ, Gordon A, deVeber
G; International Pediatric Stroke Study Group. Antithrombotic
treatments, outcomes, and prognostic factors in acute childhood-onset arterial ischaemic stroke: a multicentre, observational, cohort study. Lancet Neurol. 2009;8(12):1120-1127.
Eleftheriou D, Ganesan V. Controversies in childhood arterial ischemic stroke and cerebral venous sinus thrombosis.
Expert Rev Cardiovasc Ther. 2009;7(7):853-861.
Strter R, Kurnik K, Heller C, Schobess R, Luigs P, NowakGttl U. Aspirin versus low-dose low-molecular-weight
heparin: antithrombotic therapy in pediatric ischemic
stroke patients: a prospective follow-up study. Stroke.
2001;32(11):2554-2558.
Soman T, Rafay MF, Hune S, Allen A, MacGregor D,
deVeber G. The risks and safety of clopidogrel in pediatric arterial ischemic stroke. Stroke. 2006;37(4):1120-1122.
Bourdial H, Sassolas F, Ville D, Di Filippo S. Intravenous
thrombolysis in pediatric arterial ischemic stroke: a case
report and a review of the literature [in French]. Arch
Pediatr. 2008;15(10):1541-1546.
Ortiz GA, Koch S, Wallace DM, Lopez-Alberola R.
Successful intravenous thrombolysis for acute stroke in a
child. J Child Neurol. 2007;22(6):749-752.
Ganesan V, Prengler M, McShane MA, Wade AM, Kirkham FJ.
Investigation of risk factors in children with arterial ischemic
stroke. Ann Neurol. 2003;53(2):167-173.
Hartfield DS, Lowry NJ, Keene DL, Yager JY. Iron deficiency: a cause of stroke in infants and children. Pediatr
Neurol. 1997;16(1):50-53.
CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e799S

712. Maguire JL, deVeber G, Parkin PC. Association between


iron-deficiency anemia and stroke in young children.
Pediatrics. 2007;120(5):1053-1057.
713. Cardo E, Monrs E, Colom C, et al. Children with
stroke: polymorphism of the MTHFR gene, mild hyperhomocysteinemia, and vitamin status. J Child Neurol.
2000;15(5):295-298.
714. Prengler M, Sturt N, Krywawych S, Surtees R, Liesner R,
Kirkham F. Homozygous thermolabile variant of the methylenetetrahydrofolate reductase gene: a potential risk factor
for hyperhomocysteinaemia, CVD, and stroke in childhood.
Dev Med Child Neurol. 2001;43(4):220-225.
715. Sirachainan N, Tapanapruksakul P, Visudtibhan A, et al.
Homocysteine, MTHFR C677 T, vitamin B12, and folate
levels in Thai children with ischemic stroke: a case-control
study. J Pediatr Hematol Oncol. 2006;28(12):803-808.
716. van Beynum IM, Smeitink JA, den Heijer M, te Poele Pothoff
MT, Blom HJ. Hyperhomocysteinemia: a risk factor for
ischemic stroke in children. Circulation. 1999;99(16):
2070-2072.
717. Bonduel M, Sciuccati G, Hepner M, Torres AF, Pieroni G,
Frontroth JP. Prethrombotic disorders in children with
arterial ischemic stroke and sinovenous thrombosis. Arch
Neurol. 1999;56(8):967-971.
718. Haywood S, Liesner R, Pindora S, Ganesan V. Thrombophilia
and first arterial ischaemic stroke: a systematic review. Arch
Dis Child. 2005;90(4):402-405.
719. Abram HS, Knepper LE, Warty VS, Painter MJ. Natural history, prognosis, and lipid abnormalities of idiopathic ischemic
childhood stroke. J Child Neurol. 1996;11(4):276-282.
720. Barnes C, Newall F, Furmedge J, Mackay M, Monagle P.
Arterial ischaemic stroke in children. J Paediatr Child
Health. 2004;40(7):384-387.
721. Brankovic-Sreckovic V, Milic-Rasic V, Jovic N, Milic N,
Todorovic S. The recurrence risk of ischemic stroke in childhood. Med Princ Pract. 2004;13(3):153-158.
722. Chabrier S, Husson B, Lasjaunias P, Landrieu P, Tardieu M.
Stroke in childhood: outcome and recurrence risk by mechanism in 59 patients. J Child Neurol. 2000;15(5):290-294.
723. Chung B, Wong V. Pediatric stroke among Hong Kong
Chinese subjects. Pediatrics. 2004;114(2):e206-e212.
724. De Schryver EL, Kappelle LJ, Jennekens-Schinkel A,
Boudewyn Peters AC. Prognosis of ischemic stroke in childhood: a long-term follow-up study. Dev Med Child Neurol.
2000;42(5):313-318.
725. Ganesan V, Prengler M, Wade A, Kirkham FJ. Clinical and
radiological recurrence after childhood arterial ischemic
stroke. Circulation. 2006;114(20):2170-2177.
726. Lanthier S, Carmant L, David M, Larbrisseau A, de Veber G.
Stroke in children: the coexistence of multiple risk
factors predicts poor outcome. Neurology. 2000;54(2):
371-378.
727. Mancini J, Girard N, Chabrol B, et al. Ischemic cerebrovascular disease in children: retrospective study of 35 patients.
J Child Neurol. 1997;12(3):193-199.
728. Riikonen R, Santavuori P. Hereditary and acquired risk factors
for childhood stroke. Neuropediatrics. 1994;25(5):227-233.
729. Steinlin M, Roellin K, Schroth G. Long-term follow-up
after stroke in childhood. Eur J Pediatr. 2004;163(4-5):
245-250.
730. Agnetti A, Carano N, Sani E, et al. Cryptogenic stroke in children: possible role of patent foramen ovale. Neuropediatrics.
2006;37(1):53-56.
731. Bartz PJ, Cetta F, Cabalka AK, et al. Paradoxical emboli
in children and young adults: role of atrial septal defect
and patent foramen ovale device closure. Mayo Clin Proc.
2006;81(5):615-618.
e800S

732. Kerut EK, Norfleet WT, Plotnick GD, Giles TD. Patent
foramen ovale: a review of associated conditions and the
impact of physiological size. J Am Coll Cardiol. 2001;
38(3):613-623.
733. Rafay MF, Armstrong D, Deveber G, Domi T, Chan A,
MacGregor DL. Craniocervical arterial dissection in children: clinical and radiographic presentation and outcome. J
Child Neurol. 2006;21(1):8-16.
734. Fullerton HJ, Johnston SC, Smith WS. Arterial dissection
and stroke in children. Neurology. 2001;57(7):1155-1160.
735. Ganesan V, Chong WK, Cox TC, Chawda SJ, Prengler M,
Kirkham FJ. Posterior circulation stroke in childhood: risk
factors and recurrence. Neurology. 2002;59(10):1552-1556.
736. Lyrer P, Engelter S. Antithrombotic drugs for carotid artery
dissection. Cochrane Database Syst Rev. 2000;(4):CD000255.
737. Beletsky V, Nadareishvili Z, Lynch J, Shuaib A, Woolfenden A,
Norris JW; Canadian Stroke Consortium. Cervical arterial
dissection: time for a therapeutic trial? Stroke. 2003;34(12):
2856-2860.
738. Sbire G. Transient cerebral arteriopathy in childhood.
Lancet. 2006;368(9529):8-10.
739. Strter R, Becker S, von Eckardstein A, et al. Prospective
assessment of risk factors for recurrent stroke during
childhooda 5-year follow-up study. Lancet. 2002;360(9345):
1540-1545.
740. Benseler SM, Silverman E, Aviv RI, et al. Primary central
nervous system vasculitis in children. Arthritis Rheum.
2006;54(4):1291-1297.
741. Danchaivijitr N, Cox TC, Saunders DE, Ganesan V.
Evolution of cerebral arteriopathies in childhood arterial
ischemic stroke. Ann Neurol. 2006;59(4):620-626.
742. Ikezaki K. Rational approach to treatment of moyamoya
disease in childhood. J Child Neurol. 2000;15(5):350-356.
743. Scott RM, Smith JL, Robertson RL, Madsen JR, Soriano SG,
Rockoff MA. Long-term outcome in children with moyamoya
syndrome after cranial revascularization by pial synangiosis.
J Neurosurg. 2004;100(2)(suppl pediatrics):142-149.
744. Fung LW, Thompson D, Ganesan V. Revascularisation surgery for paediatric moyamoya: a review of the literature.
Childs Nerv Syst. 2005;21(5):358-364.
745. Scott RM. Moyamoya syndrome: a surgically treatable
cause of stroke in the pediatric patient. Clin Neurosurg.
2000;47:378-384.
746. Pegelow CH, Macklin EA, Moser FG, et al. Longitudinal
changes in brain magnetic resonance imaging findings
in children with sickle cell disease. Blood. 2002;99(8):
3014-3018.
747. Balkaran B, Char G, Morris JS, Thomas PW, Serjeant BE,
Serjeant GR. Stroke in a cohort of patients with homozygous sickle cell disease. J Pediatr. 1992;120(3):360-366.
748. Fatunde OJ, Adamson FG, Ogunseyinde O, Sodeinde O,
Familusi JB. Stroke in Nigerian children with sickle cell
disease. Afr J Med Med Sci. 2005;34(2):157-160.
749. Moohr JW, Wilson H, Pang EJ. Strokes and their management in sickle cell disease. In: Fried W, ed. Comparitive
Clinical Aspects of Sickle Cell Disease. Amsterdam, The
Netherlands: Elsevier; 1982:101-111.
750. Njamnshi AK, Mbong EN, Wonkam A, et al. The epidemiology of stroke in sickle cell patients in Yaounde, Cameroon.
J Neurol Sci. 2006;250(1-2):79-84.
751. Portnoy BA, Herion JC. Neurological manifestations in
sickle-cell disease, with a review of the literature and
emphasis on the prevalence of hemiplegia. Ann Intern Med.
1972;76(4):643-652.
752. Powars D, Wilson B, Imbus C, Pegelow C, Allen J. The
natural history of stroke in sickle cell disease. Am J Med.
1978;65(3):461-471.
Antithrombotic Therapy in Neonates and Children

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

753. Russell MO, Goldberg HI, Hodson A, et al. Effect of


transfusion therapy on arteriographic abnormalities and
on recurrence of stroke in sickle cell disease. Blood.
1984;63(1):162-169.
754. Wilimas J, Goff JR, Anderson HR Jr, Langston JW, Thompson E.
Efficacy of transfusion therapy for one to two years in
patients with sickle cell disease and cerebrovascular accidents. J Pediatr. 1980;96(2):205-208.
755. Wood DH. Cerebrovascular complications of sickle cell
anemia. Stroke. 1978;9(1):73-75.
756. Adams RJ, McKie VC, Carl EM, et al. Long-term stroke risk
in children with sickle cell disease screened with transcranial Doppler. Ann Neurol. 1997;42(5):699-704.
757. Adams RJ, McKie VC, Hsu L, et al. Prevention of a first
stroke by transfusions in children with sickle cell anemia and
abnormal results on transcranial Doppler ultrasonography.
N Engl J Med. 1998;339(1):5-11.
758. Adams RJ, Brambilla D; Optimizing Primary Stroke Prevention in Sickle Cell Anemia (STOP 2) Trial Investigators.
Discontinuing prophylactic transfusions used to prevent
stroke in sickle cell disease. N Engl J Med. 2005;353(26):
2769-2778.
759. Gulbis B, Haberman D, Dufour D, et al. Hydroxyurea for
sickle cell disease in children and for prevention of cerebrovascular events: the Belgian experience. Blood. 2005;
105(7):2685-2690.
760. Bernaudin F. Results and current indications of bone
marrow allograft in sickle cell disease [in French]. Pathol
Biol (Paris). 1999;47(1):59-64.

www.chestpubs.org

761. Vermylen C, Cornu G, Ferster A, et al. Haematopoietic


stem cell transplantation for sickle cell anaemia: the first
50 patients transplanted in Belgium. Bone Marrow Transplant.
1998;22(1):1-6.
762. Walters MC, Storb R, Patience M, et al. Impact of bone marrow
transplantation for symptomatic sickle cell disease: an interim
report. Multicenter investigation of bone marrow transplantation for sickle cell disease. Blood. 2000;95(6):1918-1924.
763. Fryer RH, Anderson RC, Chiriboga CA, Feldstein NA.
Sickle cell anemia with moyamoya disease: outcomes after
EDAS procedure. Pediatr Neurol. 2003;29(2):124-130.
764. Mendelowitsch A, Sekhar LN, Clemente R, Shuaib A.
EC-IC bypass improves chronic ischemia in a patient with
moyamoya disease secondary to sickle cell disease: an in vivo
microdialysis study. Neurol Res. 1997;19(1):66-70.
765. Schmugge M, Frischknecht H, Yonekawa Y, Baumgartner RW,
Boltshauser E, Humbert J. Stroke in hemoglobin (SD) sickle
cell disease with moyamoya: successful hydroxyurea treatment after cerebrovascular bypass surgery. Blood. 2001;
97(7):2165-2167.
766. Vernet O, Montes JL, OGorman AM, Baruchel S, Farmer JP.
Encephaloduroarterio-synangiosis in a child with sickle
cell anemia and moyamoya disease. Pediatr Neurol. 1996;
14(3):226-230.
767. National Institutes of Health; National Heart, Lung, and
Blood Institute; Division of Blood Disease and Resources.
The Management of Sickle Cell Disease. 4th ed. NIH publication no. 02-2117). Bethesda, MD: National Institutes of
Health; revised June 2002.

CHEST / 141 / 2 / FEBRUARY, 2012 SUPPLEMENT

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

e801S

CHEST

Online Data Supplement

Antithrombotic Therapy in Neonates


and Children
Antithrombotic Therapy and Prevention of Thrombosis, 9th ed:
American College of Chest Physicians Evidence-Based Clinical
Practice Guidelines
Paul Monagle, MBBS, MD, FCCP; Anthony K. C. Chan, MBBS;
Neil A. Goldenberg, MD, PhD; Rebecca N. Ichord, MD;
Janna M. Journeycake, MD, MSCS; Ulrike Nowak-Gttl, MD;
and Sara K. Vesely, PhD

2012 American College of Chest Physicians. Reproduction of this article is prohibited without written permission from the American
College of Chest Physicians
(www.chestjournal
.org/site/misc/reprints.xhtml).atDOI:
10.1378/chest.11-2308
Downloaded
from chestjournal.chestpubs.org
HINARI
on February 22, 2012

2012 American College of Chest Physicians

2012 American College of Chest Physicians. Reproduction of this article is prohibited without written permission from the American
College of Chest Physicians
(www.chestjournal.org/site/misc/reprints.xhtml).
10.1378/chest.11-2308
Downloaded
from chestjournal.chestpubs.org atDOI:
HINARI
on February 22, 2012

2012 American College of Chest Physicians

Retrospective
cohort

Retrospective
cohort

Case series

Case series

RCT

Cohort

Biss et al2/2008

Goldenberg et al3/2005

Revel Vilk et al4/2004

Massicotte et al5/2003

Streif et al6/2003

Design

Blatn and
Fiamoli1/2009

Study/Year

LMWH (anti-Xa
0.5-1.0 units/mL)

UFH/VKA
(INR 2-3)
46% in TTR
Reviparin
(anti-Xa
0.5-1.0 units/mL)
75% in range

Enoxaparin
(anti-Xa
0.5-1.0 units/mL)
Duration 5-616 d
enoxaparin
Previous
anticoagulation,
lytic therapy,
or both

LMWH (anti-Xa
0.5-1.2 units/mL)
VKA (INR 2-3)

UFH (anti-Xa
0.35-0.7 units/mL)
LMWH (anti-Xa
0.5-1.0 units/mL)
VKA (INR 2.5),
(range, 2-3)

LMWH (anti-Xa
0.5-1.0 units/mL)

Treatment Target
Levels

N 5 62

Reviparin, n 5 36

N 5 78, N 5 76 evaluable
UFH/VKA, n 5 40

N 5 245
Age, 33 d-17.3 y
56% neonates
VTE, 84%
Arterial TE, 5%

N 5 11
LMWH, n 5 4
VKA, n 5 1
Lemierre
syndrome, n 5 6

Pulmonary embolus,
N 5 56
UFH, n 5 36
Thrombolysis, n 5 8
IVC filter, n 5 7
Thrombectomy, n 5 2

N 5 33
IV LMWH, n 5 21
Subcutaneous
LMWH, n 5 12

No. Patients

1.2/100 treatment-y

3 mo therapy
plus 3 mo
follow-up

Mean, 3 d-6.6 y
(median, 6 m);
190 had
evaluable
ultrasounds

Median, 1 y (range,
7 wk-12 mo)

Median, 56 mo
Range 5-93 mo

Not stated

Follow-up

n54

2 (5.6%)

2 (5.6%)

n53

5 (12.5%)

No reported
events

No reported
events

n 5 12
Fatal bleeding
n52

Major
Hemorrhage

4 (10%)

Developed PE,
n55

No reported
events

Early progression,
n 54
Recurrence, n 5 7
Fatal thrombosis,
n 55

Thromboembolic
Events

Table S1[Recommendation 2.22.1] Evidence Profile: Studies Assessing Anticoagulation for VTE in Children

(Continued)

Underpowered

22% had no serial


ultrasounds to
assess thrombus
following treatment
Collaterals, 20%
Clot resolution, 53%
No change, 34%
Partial resolution, 25%

At follow-up 4-6 mo
postdiagnosis
n 5 2 persistent
thrombosis
n 5 4 recanalized

Overall mortality, 21%

Full or partial
recanalization:
85% for IV LMWH,
66% for
subcutaneous
LMWH

Comments

2012 American College of Chest Physicians. Reproduction of this article is prohibited without written permission from the American
College of Chest Physicians
(www.chestjournal.org/site/misc/reprints.xhtml).
10.1378/chest.11-2308
Downloaded
from chestjournal.chestpubs.org atDOI:
HINARI
on February 22, 2012

2012 American College of Chest Physicians

Retrospective
case series

Case series

Case series

Case series

Dix et al8/2000

Punzalan et al9/2000

Nohe et al10/1999

Design

Hofmann et al7/2001

Study/Year

N5 19
Prospective, n 5 6
Retrospective, n 5 13
VTE, n 5 14
Primary prophylaxis,
n55
N 5 48
VTE, n 5 19

Dalteparin 129
43 units/kg per d
(anti-Xa 0.4-1.0 units/mL)

N 5 138 (146 courses


of therapy)
VTE, n 5 86
Age, 1 d-18 y
Median, 3.5 y
Mean, 6.1 y

n 5 66
n 5 13
48 girls
31 boys
Aged 2 wk-19 y,
Mean, 11.8 y

No. Patients

Enoxaparin
(anti-Xa 0.5-1.2 units/mL)
Enoxaparin or lytic
therapy

Enoxaparin (anti-Xa
0.5-1.0 units/mL)
Low-risk prophylaxis (anti-Xa
0.1-0.4 units/mL) VKA
added in 47 of 146

Nadroparin
Enoxaparin (anti-Xa
0.5-1.0 units/mL)

Treatment Target
Levels

Not reported

Not reported

Duration of therapy:
n 5 74 of
146 20 d
treatment
Not reported

N 5 62 short-term
prophylaxis
(1-2 wk)
N 5 13 (long-term
therapy and
follow-up)

Follow-up

Table S1Continued

None reported

No new/recurrent
VTE

n 5 2 (1.3%)
recurrent/new
VTE when patient
subtherapeutic

n51

Thromboembolic
Events

None reported

No major
hemorrhage

n 5 7 (5%) major

n50

Major
Hemorrhage

(Continued)

n 5 6 complete
recanalization
n 5 5 partial
recanalization
n 5 7 no recanalization

Unable to determine
which patients were
treated for VTE
vs primary
prophylaxis

n 5 12 lost to
follow-up;
transferred to other
hospitals and two
deaths directly related
to therapy (one
hemorrhage; one TE)

n 5 4 no reocclusion
n 5 2 recanalized
n 5 1 reocclusion
Unable to separate
which patients
received enoxaparin
vs nadroparin

Comments

2012 American College of Chest Physicians. Reproduction of this article is prohibited without written permission from the American
College of Chest Physicians
(www.chestjournal.org/site/misc/reprints.xhtml).
10.1378/chest.11-2308
Downloaded
from chestjournal.chestpubs.org atDOI:
HINARI
on February 22, 2012

2012 American College of Chest Physicians

Case series

Prospective
cohort

Prospective
cohort

Massicotte et al12/1996

Andrew et al13/1994

Andrew et al14/1994
VKA
1. INR 2-3 3 3-6 mo
followed by
INR 1.3-1.8 secondary
prophylaxis
2. 1.3-1.8 primary
prophylaxis
3. 2.5-3.5
(mechanical valves)

UFH (PTT 55-85 s);


52/65 (73%) of
PTTs corresponded
to anti-Xa and within
therapeutic range

Enoxaparin (anti-Xa
0.5-1.0 units/mL)

VKA
n 5 52, (INR 1.4-1.8)
n 5 263, (INR 2-3)
n 5 (INR 2.5-3.5)
n 5 208 primary
prophylaxis
n 5 144 secondary
prophylaxis
49-61% TTR

Treatment Target
Levels

N 5 115
Secondary prophylaxis,
n 5 54
Primary prophylaxis,
n 5 61

N 5 65
Follow-up, N 5 65
n 5 29, , 1 y
n 5 36, 1-18 y
VTE, n 5 30
(n 5 9 , 1 y)
Arterial thrombosis,
n 5 11

N 5 25
VTE, n 5 14
CNS, n 5 9
CHD prophylaxis,
n52

N 5 319
352 courses of
therapy
391 treatment-y
Age, 1 mo-18 y

No. Patients

Duration of therapy
n 5 37, 3-6 mo
n 5 38, 6 mo
n 5 40,
lifelong
18 mo

Not reported

Variable duration
of therapy
Range, 10-50 d

Median, 6 mo

Follow-up

n 5 4 new/recurrent
thrombosis after
VKA therapy
discontinued

n52

No new/recurrent
VTE

n58
n 5 6, after VKA
therapy
discontinued
n52
1.3% per patient-y,
during VKA
therapy

Thromboembolic
Events

n52

n51

n52

n52
0.5%/patient-y

Major
Hemorrhage

36% of children had


CHD (n 5 114)

Comments

CHD 5 congenital heart disease; INR 5 international normalized ratio; LMWH 5 low-molecular-weight heparin; PTT 5 partial thromboplastin time; TE 5 thromboembolism; TTR 5 time in therapeutic
range; UFH 5 unfractionated heparin; VKA 5 vitamin K antagonist.

11

Prospective
cohort

Design

Streif et al /1999

Study/Year

Table S1Continued

References
1. Blatn J, Fiamoli V. Treatment of deep vein thrombosis with
continuous intravenous infusion of LMWH in childrenan
alternative to subcutaneous application when needed. Vnitr
Lek. 2009;55(3):227-232.
2. Biss TT, Brando LR, Kahr WH, Chan AK, Williams S.
Clinical features and outcome of pulmonary embolism in
children. Br J Haematol. 2008;142(5):808-818.
3. Goldenberg NA, Knapp-Clevenger R, Hays T, MancoJohnson MJ. Lemierres and Lemierres-like syndromes in
children: survival and thromboembolic outcomes. Pediatrics.
2005;116(4):e543-e548.
4. Revel-Vilk S, Sharathkumar A, Massicotte P, et al. Natural
history of arterial and venous thrombosis in children treated
with low molecular weight heparin: a longitudinal study by
ultrasound. J Thromb Haemost. 2004;2(1):42-46.
5. Massicotte P, Julian JA, Gent M, et al. An open-label randomized controlled trial of low molecular weight heparin compared
to heparin and coumadin for the treatment of venous thromboembolic events in children: the REVIVE trial. Thromb Res.
2003;109:85-92.
6. Streif W, Goebel G, Chan AK, Massicotte MP. Use of low
molecular mass heparin (enoxaparin) in newborn infants: a
prospective cohort study of 62 patients. Arch Dis Child Fetal
Neonatal Ed. 2003;88(5):F365-F370.

7. Hofmann S, Knoefler R, Lorenz N, et al. Clinical experiences


with low-molecular weight heparins in pediatric patients.
Thromb Res. 2001;103:345-353.
8. Dix D, Andrew M, Marzinotto V, et al. The use of low molecular weight heparin in pediatric patients: a prospective cohort
study. J Pediatr. 2000;136(4):439-445.
9. Punzalan RC, Hillery CA, Montgomery RR, Scott CA, Gill JC.
Low-molecular-weight heparin in thrombotic disease in children
and adolescents. J Pediatr Hematol Oncol. 2000;22(2):137-142.
10. Nohe N, Flemmer A, Rmler R, Praun M, Auberger K.
The low molecular weight heparin dalteparin for prophylaxis
and therapy of thrombosis in childhood: a report on 48 cases.
Eur J Pediatr. 1999;158(suppl 3):S134-S139.
11. Streif W, Andrew M, Marzinotto V, et al. Analysis of warfarin
therapy in pediatric patients: A prospective cohort study of
319 patients. Blood. 1999;94(9):3007-3014.
12. Massicotte P, Adams M, Marzinotto V, Brooker LA, Andrew M.
Low-molecular-weight heparin in pediatric patients with
thrombotic disease: a dose finding study. J Pediatr. 1996;
128(3):313-318.
13. Andrew M, Marzinotto V, Massicotte P, et al. Heparin therapy
in pediatric patients: a prospective cohort study. Pediatr Res.
1994;35(1):78-83.
14. Andrew M, Marzinotto V, Brooker LA, et al. Oral anticoagulation therapy in pediatric patients: a prospective study.
Thromb Haemost. 1994;71:265-269.

2012 American College of Chest Physicians. Reproduction of this article is prohibited without written permission from the American
College of Chest Physicians
(www.chestjournal.org/site/misc/reprints.xhtml).
10.1378/chest.11-2308
Downloaded
from chestjournal.chestpubs.org atDOI:
HINARI
on February 22, 2012

2012 American College of Chest Physicians

Antithrombotic Therapy in Neonates and Children : Antithrombotic


Therapy and Prevention of Thrombosis, 9th ed: American College of
Chest Physicians Evidence-Based Clinical Practice Guidelines
Paul Monagle, Anthony K. C. Chan, Neil A. Goldenberg, Rebecca N. Ichord,
Janna M. Journeycake, Ulrike Nowak-Gttl and Sara K. Vesely
Chest 2012;141; e737S-e801S
DOI 10.1378/chest.11-2308
This information is current as of February 22, 2012
Supplementary Material
View e-supplements related to this article at:
http://chestjournal.chestpubs.org/content/suppl/2012/02/03/141.2_suppl.e737S.DC1.html
Updated Information & Services
Updated Information and services can be found at:
http://chestjournal.chestpubs.org/content/141/2_suppl/e737S.full.html
References
This article cites 750 articles, 208 of which can be accessed free at:
http://chestjournal.chestpubs.org/content/141/2_suppl/e737S.full.html#ref-list-1
Cited Bys
This article has been cited by 2 HighWire-hosted articles:
http://chestjournal.chestpubs.org/content/141/2_suppl/e737S.full.html#related-urls
Permissions & Licensing
Information about reproducing this article in parts (figures, tables) or in its entirety can be
found online at:
http://www.chestpubs.org/site/misc/reprints.xhtml
Reprints
Information about ordering reprints can be found online:
http://www.chestpubs.org/site/misc/reprints.xhtml
Citation Alerts
Receive free e-mail alerts when new articles cite this article. To sign up, select the
"Services" link to the right of the online article.
Images in PowerPoint format
Figures that appear in CHEST articles can be downloaded for teaching purposes in
PowerPoint slide format. See any online figure for directions.

Downloaded from chestjournal.chestpubs.org at HINARI on February 22, 2012


2012 American College of Chest Physicians

You might also like